Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38979307

ABSTRACT

Nucleosomes are the fundamental unit of eukaryotic chromatin. Diverse factors interact with nucleosomes to modulate chromatin architecture and facilitate DNA repair, replication, transcription, and other cellular processes. An important platform for chromatin binding is the H2A-H2B acidic patch. Here, we used AlphaFold-Multimer to screen over 7000 human proteins for nucleosomal acidic patch binding and identify 41 potential acidic patch binders. We determined the cryo-EM structure of one hit, SHPRH, with the nucleosome at 2.8 Å. The structure confirms the predicted acidic patch interaction, reveals that the SHPRH ATPase engages a different nucleosomal DNA location than other SF2-type ATPases, and clarifies the roles of SHPRH's domains in nucleosome recognition. Our results illustrate the use of in silico screening as a high throughput method to identify specific interaction types and expands the set of potential acidic patch binding factors. All the screening data is freely available at: https://predictomes.org/view/acidicpatch.

2.
Science ; : eado3867, 2024 06 20.
Article in English | MEDLINE | ID: mdl-38900911

ABSTRACT

Using CRISPR/Cas9 nicking enzymes, we examine the interaction between the replication machinery and single strand breaks, one of the most common forms of endogenous DNA damage. We show that replication fork collapse at leading strand nicks generates resected single-ended double-strand breaks (seDSBs) that are repaired by homologous recombination (HR). If these seDSBs are not promptly repaired, arrival of adjacent forks creates double ended DSBs (deDSBs), which could drive genomic scarring in HR-deficient cancers. deDSBs can also be generated directly when the replication fork bypasses lagging strand nicks. Unlike deDSBs produced independently of replication, end-resection at nick-induced se/deDSBs is BRCA1-independent. Nevertheless, BRCA1 antagonizes 53BP1 suppression of RAD51 filament formation. These results highlight unique mechanisms that maintain replication fork stability.

3.
bioRxiv ; 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38645019

ABSTRACT

Protein-protein interactions (PPIs) are ubiquitous in biology, yet a comprehensive structural characterization of the PPIs underlying biochemical processes is lacking. Although AlphaFold-Multimer (AF-M) has the potential to fill this knowledge gap, standard AF-M confidence metrics do not reliably separate relevant PPIs from an abundance of false positive predictions. To address this limitation, we used machine learning on well curated datasets to train a Structure Prediction and Omics informed Classifier called SPOC that shows excellent performance in separating true and false PPIs, including in proteome-wide screens. We applied SPOC to an all-by-all matrix of nearly 300 human genome maintenance proteins, generating ~40,000 predictions that can be viewed at predictomes.org, where users can also score their own predictions with SPOC. High confidence PPIs discovered using our approach suggest novel hypotheses in genome maintenance. Our results provide a framework for interpreting large scale AF-M screens and help lay the foundation for a proteome-wide structural interactome.

4.
Nat Commun ; 15(1): 1250, 2024 Feb 10.
Article in English | MEDLINE | ID: mdl-38341432

ABSTRACT

Nonhomologous end joining (NHEJ), the primary pathway of vertebrate DNA double-strand-break (DSB) repair, directly re-ligates broken DNA ends. Damaged DSB ends that cannot be immediately re-ligated are modified by NHEJ processing enzymes, including error-prone polymerases and nucleases, to enable ligation. However, DSB ends that are initially compatible for re-ligation are typically joined without end processing. As both ligation and end processing occur in the short-range (SR) synaptic complex that closely aligns DNA ends, it remains unclear how ligation of compatible ends is prioritized over end processing. In this study, we identify structural interactions of the NHEJ-specific DNA Ligase IV (Lig4) within the SR complex that prioritize ligation and promote NHEJ fidelity. Mutational analysis demonstrates that Lig4 must bind DNA ends to form the SR complex. Furthermore, single-molecule experiments show that a single Lig4 binds both DNA ends at the instant of SR synapsis. Thus, Lig4 is poised to ligate compatible ends upon initial formation of the SR complex before error-prone processing. Our results provide a molecular basis for the fidelity of NHEJ.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Ligase ATP/metabolism , DNA Repair , DNA Ligases/metabolism , DNA/genetics , DNA/metabolism
5.
Mol Cell ; 84(3): 404-408, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38306999

ABSTRACT

To celebrate the 50th anniversary of Cell Press and the Cell focus issue on structural biology, we discussed with scientists working across diverse fields how AlphaFold has changed their research and brought structural biology to the masses.


Subject(s)
Anniversaries and Special Events , Molecular Biology
6.
Science ; 381(6664): eadi3448, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37590370

ABSTRACT

CDC45-MCM2-7-GINS (CMG) helicase assembly is the central event in eukaryotic replication initiation. In yeast, a multi-subunit "pre-loading complex" (pre-LC) accompanies GINS to chromatin-bound MCM2-7, leading to CMG formation. Here, we report that DONSON, a metazoan protein mutated in microcephalic primordial dwarfism, is required for CMG assembly in vertebrates. Using AlphaFold to screen for protein-protein interactions followed by experimental validation, we show that DONSON scaffolds a vertebrate pre-LC containing GINS, TOPBP1, and DNA pol ε. Our evidence suggests that DONSON docks the pre-LC onto MCM2-7, delivering GINS to its binding site in CMG. A patient-derived DONSON mutation compromises CMG assembly and recapitulates microcephalic dwarfism in mice. These results unify our understanding of eukaryotic replication initiation, implicate defective CMG assembly in microcephalic dwarfism, and illustrate how in silico protein-protein interaction screening accelerates mechanistic discovery.


Subject(s)
Cell Cycle Proteins , DNA Replication , DNA-Binding Proteins , Minichromosome Maintenance Proteins , Nuclear Proteins , Animals , Humans , Mice , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Minichromosome Maintenance Proteins/genetics , Minichromosome Maintenance Proteins/metabolism , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins , Protein Interaction Mapping/methods , Computer Simulation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Dwarfism/genetics , Microcephaly/genetics , Xenopus laevis
7.
Cell Rep ; 42(2): 112125, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36807144

ABSTRACT

Tripartite motif-containing protein 21 (TRIM21) is a cytosolic antibody receptor and E3 ubiquitin ligase that promotes destruction of a broad range of pathogens. TRIM21 also underlies the antibody-dependent protein targeting method Trim-Away. Current evidence suggests that TRIM21 binding to antibodies leads to formation of a self-anchored K63 ubiquitin chain on the N terminus of TRIM21 that triggers the destruction of TRIM21, antibody, and target protein. Here, we report that addition of antibody and TRIM21 to Xenopus egg extracts promotes efficient degradation of endogenous target proteins, establishing cell-free Trim-Away as a powerful tool to interrogate protein function. Chemical methylation of TRIM21 had no effect on target proteolysis, whereas deletion of all lysine residues in targets abolished their ubiquitination and proteasomal degradation. These results demonstrate that target protein, but not TRIM21, polyubiquitination is required for Trim-Away, and they suggest that current models of TRIM21 function should be fundamentally revised.


Subject(s)
Proteins , Ubiquitin-Protein Ligases , Ubiquitination , Ubiquitin-Protein Ligases/metabolism , Proteins/metabolism , Ubiquitin/metabolism , Antibodies/metabolism
8.
Mol Cell ; 83(1): 43-56.e10, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36608669

ABSTRACT

Endogenous and exogenous agents generate DNA-protein crosslinks (DPCs), whose replication-dependent degradation by the SPRTN protease suppresses aging and liver cancer. SPRTN is activated after the replicative CMG helicase bypasses a DPC and polymerase extends the nascent strand to the adduct. Here, we identify a role for the 5'-to-3' helicase FANCJ in DPC repair. In addition to supporting CMG bypass, FANCJ is essential for SPRTN activation. FANCJ binds ssDNA downstream of the DPC and uses its ATPase activity to unfold the protein adduct, which exposes the underlying DNA and enables cleavage of the adduct. FANCJ-dependent DPC unfolding is also essential for translesion DNA synthesis past DPCs that cannot be degraded. In summary, our results show that helicase-mediated protein unfolding enables multiple events in DPC repair.


Subject(s)
DNA Damage , DNA-Binding Proteins , Protein Unfolding , DNA/genetics , DNA/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair , DNA Replication , DNA-Binding Proteins/genetics
9.
Nat Commun ; 13(1): 6591, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36329031

ABSTRACT

The p97 ATPase extracts polyubiquitylated proteins from diverse cellular structures in preparation for destruction by the proteasome. p97 functions with Ufd1-Npl4 and a variety of UBA-UBX co-factors, but how p97 complexes assemble on ubiquitylated substrates is unclear. To address this, we investigated how p97 disassembles the CMG helicase after it is ubiquitylated during replication termination. We show that p97Ufd1-Npl4 recruitment to CMG requires the UBA-UBX protein Ubxn7, and conversely, stable Ubxn7 binding to CMG requires p97Ufd1-Npl4. This cooperative assembly involves interactions between Ubxn7, p97, Ufd1-Npl4, and ubiquitin. Another p97 co-factor, Faf1, partially compensates for the loss of Ubxn7. Surprisingly, p97Ufd1-Npl4-Ubxn7 and p97Ufd1-Npl4-Faf1 also assemble cooperatively on unanchored ubiquitin chains. We propose that cooperative and substrate-independent recognition of ubiquitin chains allows p97 to recognize an unlimited number of polyubiquitylated proteins while avoiding the formation of partial, inactive complexes.


Subject(s)
DNA Replication , Ubiquitin , Protein Binding , Ubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism
10.
Nat Struct Mol Biol ; 29(5): 451-462, 2022 05.
Article in English | MEDLINE | ID: mdl-35534579

ABSTRACT

The 5-hydroxymethylcytosine binding, embryonic stem-cell-specific (HMCES) protein forms a covalent DNA-protein cross-link (DPC) with abasic (AP) sites in single-stranded DNA, and the resulting HMCES-DPC is thought to suppress double-strand break formation in S phase. However, the dynamics of HMCES cross-linking and whether any DNA repair pathways normally include an HMCES-DPC intermediate remain unknown. Here, we use Xenopus egg extracts to show that an HMCES-DPC forms on the AP site generated during replication-coupled DNA interstrand cross-link repair. We show that HMCES cross-links form on DNA after the replicative CDC45-MCM2-7-GINS (CMG) helicase has passed over the AP site, and that HMCES is subsequently removed by the SPRTN protease. The HMCES-DPC suppresses double-strand break formation, slows translesion synthesis past the AP site and introduces a bias for insertion of deoxyguanosine opposite the AP site. These data demonstrate that HMCES-DPCs form as intermediates in replication-coupled repair, and they suggest a general model of how HMCES protects AP sites during DNA replication.


Subject(s)
DNA-Binding Proteins , DNA , DNA/metabolism , DNA Damage , DNA Repair , DNA Replication , DNA-Binding Proteins/metabolism
11.
Nature ; 605(7909): 357-365, 2022 05.
Article in English | MEDLINE | ID: mdl-35508654

ABSTRACT

The entry of mammalian cells into the DNA synthesis phase (S phase) represents a key event in cell division1. According to current models of the cell cycle, the kinase CDC7 constitutes an essential and rate-limiting trigger of DNA replication, acting together with the cyclin-dependent kinase CDK2. Here we show that CDC7 is dispensable for cell division of many different cell types, as determined using chemical genetic systems that enable acute shutdown of CDC7 in cultured cells and in live mice. We demonstrate that another cell cycle kinase, CDK1, is also active during G1/S transition both in cycling cells and in cells exiting quiescence. We show that CDC7 and CDK1 perform functionally redundant roles during G1/S transition, and at least one of these kinases must be present to allow S-phase entry. These observations revise our understanding of cell cycle progression by demonstrating that CDK1 physiologically regulates two distinct transitions during cell division cycle, whereas CDC7 has a redundant function in DNA replication.


Subject(s)
Cell Cycle Proteins , G1 Phase , Protein Serine-Threonine Kinases , Proteolysis , S Phase , Animals , Cell Cycle Proteins/metabolism , DNA Replication , Mice , Protein Serine-Threonine Kinases/metabolism
12.
Nucleic Acids Res ; 49(22): 13194-13206, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34850944

ABSTRACT

When vertebrate replisomes from neighboring origins converge, the Mcm7 subunit of the replicative helicase, CMG, is ubiquitylated by the E3 ubiquitin ligase, CRL2Lrr1. Polyubiquitylated CMG is then disassembled by the p97 ATPase, leading to replication termination. To avoid premature replisome disassembly, CRL2Lrr1 is only recruited to CMGs after they converge, but the underlying mechanism is unclear. Here, we use cryogenic electron microscopy to determine structures of recombinant Xenopus laevis CRL2Lrr1 with and without neddylation. The structures reveal that CRL2Lrr1 adopts an unusually open architecture, in which the putative substrate-recognition subunit, Lrr1, is located far from the catalytic module that catalyzes ubiquitin transfer. We further demonstrate that a predicted, flexible pleckstrin homology domain at the N-terminus of Lrr1 is essential to target CRL2Lrr1 to terminated CMGs. We propose a hypothetical model that explains how CRL2Lrr1's catalytic module is positioned next to the ubiquitylation site on Mcm7, and why CRL2Lrr1 binds CMG only after replisomes converge.


Subject(s)
DNA Replication/genetics , Ubiquitin-Protein Ligases/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Animals , Cryoelectron Microscopy , DNA Helicases/genetics , DNA Helicases/metabolism , Minichromosome Maintenance Complex Component 7/genetics , Minichromosome Maintenance Complex Component 7/metabolism , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Conformation , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Sf9 Cells , Spodoptera , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/ultrastructure , Ubiquitination , Xenopus Proteins/chemistry , Xenopus Proteins/metabolism , Xenopus laevis/metabolism
13.
Nat Cell Biol ; 23(6): 595-607, 2021 06.
Article in English | MEDLINE | ID: mdl-34108663

ABSTRACT

Cells employ transcription-coupled repair (TCR) to eliminate transcription-blocking DNA lesions. DNA damage-induced binding of the TCR-specific repair factor CSB to RNA polymerase II (RNAPII) triggers RNAPII ubiquitylation of a single lysine (K1268) by the CRL4CSA ubiquitin ligase. How CRL4CSA is specifically directed towards K1268 is unknown. Here, we identify ELOF1 as the missing link that facilitates RNAPII ubiquitylation, a key signal for the assembly of downstream repair factors. This function requires its constitutive interaction with RNAPII close to K1268, revealing ELOF1 as a specificity factor that binds and positions CRL4CSA for optimal RNAPII ubiquitylation. Drug-genetic interaction screening also revealed a CSB-independent pathway in which ELOF1 prevents R-loops in active genes and protects cells against DNA replication stress. Our study offers key insights into the molecular mechanisms of TCR and provides a genetic framework of the interplay between transcriptional stress responses and DNA replication.


Subject(s)
DNA Damage , DNA Repair , Peptide Elongation Factor 1/metabolism , RNA Polymerase II/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , CRISPR-Cas Systems , Cell Line, Tumor , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Humans , Peptide Elongation Factor 1/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Protein Binding , Protein Interaction Domains and Motifs , RNA Polymerase II/genetics , Transcription Elongation, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
14.
Annu Rev Biochem ; 90: 107-135, 2021 06 20.
Article in English | MEDLINE | ID: mdl-33882259

ABSTRACT

DNA interstrand cross-links (ICLs) covalently connect the two strands of the double helix and are extremely cytotoxic. Defective ICL repair causes the bone marrow failure and cancer predisposition syndrome, Fanconi anemia, and upregulation of repair causes chemotherapy resistance in cancer. The central event in ICL repair involves resolving the cross-link (unhooking). In this review, we discuss the chemical diversity of ICLs generated by exogenous and endogenous agents. We then describe how proliferating and nonproliferating vertebrate cells unhook ICLs. We emphasize fundamentally new unhooking strategies, dramatic progress in the structural analysis of the Fanconi anemia pathway, and insights into how cells govern the choice between different ICL repair pathways. Throughout, we highlight the many gaps that remain in our knowledge of these fascinating DNA repair pathways.


Subject(s)
DNA Damage/genetics , DNA Repair/physiology , Fanconi Anemia/genetics , Vertebrates/genetics , Acetaldehyde/metabolism , Animals , DNA/chemistry , DNA Breaks, Double-Stranded , DNA Breaks, Single-Stranded , DNA Replication , Fanconi Anemia/metabolism , Humans
15.
Mol Cell ; 81(6): 1309-1318.e6, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33484638

ABSTRACT

DNA damage impedes replication fork progression and threatens genome stability. Upon encounter with most DNA adducts, the replicative CMG helicase (CDC45-MCM2-7-GINS) stalls or uncouples from the point of synthesis, yet eventually resumes replication. However, little is known about the effect on replication of single-strand breaks or "nicks," which are abundant in mammalian cells. Using Xenopus egg extracts, we reveal that CMG collision with a nick in the leading strand template generates a blunt-ended double-strand break (DSB). Moreover, CMG, which encircles the leading strand template, "runs off" the end of the DSB. In contrast, CMG collision with a lagging strand nick generates a broken end with a single-stranded overhang. In this setting, CMG translocates along double-stranded DNA beyond the break and is then ubiquitylated and removed from chromatin by the same pathway used during replication termination. Our results show that nicks are uniquely dangerous DNA lesions that invariably cause replisome disassembly, and they suggest that CMG cannot be stored on dsDNA while cells resolve replication stress.


Subject(s)
Chromatin , DNA Breaks, Single-Stranded , DNA Helicases , DNA Replication , Ubiquitination , Xenopus Proteins , Animals , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Helicases/metabolism , Sf9 Cells , Spodoptera , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis
16.
Trends Cell Biol ; 31(2): 75-85, 2021 02.
Article in English | MEDLINE | ID: mdl-33317933

ABSTRACT

In preparation for cell division, the genome must be copied with high fidelity. However, replisomes often encounter obstacles, including bulky DNA lesions caused by reactive metabolites and chemotherapeutics, as well as stable nucleoprotein complexes. Here, we discuss recent advances in our understanding of TRAIP, a replisome-associated E3 ubiquitin ligase that is mutated in microcephalic primordial dwarfism. In interphase, TRAIP helps replisomes overcome DNA interstrand crosslinks and DNA-protein crosslinks, whereas in mitosis it triggers disassembly of all replisomes that remain on chromatin. We describe a model to explain how TRAIP performs these disparate functions and how they help maintain genome integrity.


Subject(s)
DNA Helicases/metabolism , DNA Repair , DNA Replication , Mitosis , Ubiquitin-Protein Ligases/metabolism , Animals , DNA Helicases/chemistry , Humans , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Xenopus laevis
18.
Genes Dev ; 34(21-22): 1534-1545, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32943574

ABSTRACT

When converging replication forks meet during replication termination, the CMG (Cdc45-MCM2-7-GINS) helicase is polyubiquitylated by CRL2Lrr1 and unloaded from chromatin by the p97 ATPase. Here, we investigate the signal that triggers CMG unloading in Xenopus egg extracts using single-molecule and ensemble approaches. We show that converging CMGs pass each other and keep translocating at the same speed as before convergence, whereafter they are rapidly and independently unloaded. When CMG unloading is blocked, diverging CMGs do not support DNA synthesis, indicating that after bypass CMGs encounter the nascent lagging strands of the converging fork and then translocate along double-stranded DNA (dsDNA). However, translocation on dsDNA is not required for CMG's removal from chromatin because in the absence of nascent strand synthesis, converging CMGs are still unloaded. Moreover, recombinant CMG added to nuclear extract undergoes ubiquitylation and disassembly in the absence of any DNA, and DNA digestion triggers CMG ubiquitylation at stalled replication forks. Our findings suggest that DNA suppresses CMG ubiquitylation during elongation and that this suppression is relieved when CMGs converge, leading to CMG unloading.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Replication , Xenopus Proteins/metabolism , Animals , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , DNA/chemistry , DNA/metabolism , Minichromosome Maintenance Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ubiquitination , Xenopus laevis/genetics , Xenopus laevis/metabolism
19.
Mol Cell ; 79(2): 221-233.e5, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32603710

ABSTRACT

Cas9 is a prokaryotic RNA-guided DNA endonuclease that binds substrates tightly in vitro but turns over rapidly when used to manipulate genomes in eukaryotic cells. Little is known about the factors responsible for dislodging Cas9 or how they influence genome engineering. Unbiased detection through proximity labeling of transient protein interactions in cell-free Xenopus laevis egg extract identified the dimeric histone chaperone facilitates chromatin transcription (FACT) as an interactor of substrate-bound Cas9. FACT is both necessary and sufficient to displace dCas9, and FACT immunodepletion converts Cas9's activity from multi-turnover to single turnover. In human cells, FACT depletion extends dCas9 residence times, delays genome editing, and alters the balance between indel formation and homology-directed repair. FACT knockdown also increases epigenetic marking by dCas9-based transcriptional effectors with a concomitant enhancement of transcriptional modulation. FACT thus shapes the intrinsic cellular response to Cas9-based genome manipulation most likely by determining Cas9 residence times.


Subject(s)
CRISPR-Associated Protein 9/metabolism , DNA-Binding Proteins/metabolism , Genome, Human , High Mobility Group Proteins/metabolism , Transcriptional Elongation Factors/metabolism , Animals , CRISPR-Associated Proteins/metabolism , Cell Line , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Epigenesis, Genetic , Gene Editing , Gene Knockdown Techniques , Humans , Nucleosomes/metabolism , Xenopus laevis
20.
Nat Commun ; 11(1): 2104, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32355176

ABSTRACT

The response to DNA damage-stalled RNA polymerase II (RNAPIIo) involves the assembly of the transcription-coupled repair (TCR) complex on actively transcribed strands. The function of the TCR proteins CSB, CSA and UVSSA and the manner in which the core DNA repair complex, including transcription factor IIH (TFIIH), is recruited are largely unknown. Here, we define the assembly mechanism of the TCR complex in human isogenic knockout cells. We show that TCR is initiated by RNAPIIo-bound CSB, which recruits CSA through a newly identified CSA-interaction motif (CIM). Once recruited, CSA facilitates the association of UVSSA with stalled RNAPIIo. Importantly, we find that UVSSA is the key factor that recruits the TFIIH complex in a manner that is stimulated by CSB and CSA. Together these findings identify a sequential and highly cooperative assembly mechanism of TCR proteins and reveal the mechanism for TFIIH recruitment to DNA damage-stalled RNAPIIo to initiate repair.


Subject(s)
Carrier Proteins/metabolism , DNA Damage , DNA Helicases/metabolism , DNA Repair Enzymes/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Polymerase II/metabolism , Transcription Factor TFIIH/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , DNA Repair , Humans , Transcription, Genetic , Ultraviolet Rays , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...