Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 14(1): 341, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36670096

ABSTRACT

The transcriptional response to genotoxic stress involves gene expression arrest, followed by recovery of mRNA synthesis (RRS) after DNA repair. We find that the lack of the EXD2 nuclease impairs RRS and decreases cell survival after UV irradiation, without affecting DNA repair. Overexpression of wild-type, but not nuclease-dead EXD2, restores RRS and cell survival. We observe that UV irradiation triggers the relocation of EXD2 from mitochondria to the nucleus. There, EXD2 is recruited to chromatin where it transiently interacts with RNA Polymerase II (RNAPII) to promote the degradation of nascent mRNAs synthesized at the time of genotoxic attack. Reconstitution of the EXD2-RNAPII partnership on a transcribed DNA template in vitro shows that EXD2 primarily interacts with an elongation-blocked RNAPII and efficiently digests mRNA. Overall, our data highlight a crucial step in the transcriptional response to genotoxic attack in which EXD2 interacts with elongation-stalled RNAPII on chromatin to potentially degrade the associated nascent mRNA, allowing transcription restart after DNA repair.


Subject(s)
DNA Damage , DNA Repair , Chromatin/genetics , Transcription, Genetic , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Messenger/genetics
2.
Nat Commun ; 10(1): 1288, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894545

ABSTRACT

The TFIIH subunit XPB is involved in combined Xeroderma Pigmentosum and Cockayne syndrome (XP-B/CS). Our analyses reveal that XPB interacts functionally with KAT2A, a histone acetyltransferase (HAT) that belongs to the hSAGA and hATAC complexes. XPB interacts with KAT2A-containing complexes on chromatin and an XP-B/CS mutation specifically elicits KAT2A-mediated large-scale chromatin decondensation. In XP-B/CS cells, the abnormal recruitment of TFIIH and KAT2A to chromatin causes inappropriate acetylation of histone H3K9, leading to aberrant formation of transcription initiation complexes on the promoters of several hundred genes and their subsequent overexpression. Significantly, this cascade of events is similarly sensitive to KAT2A HAT inhibition or to the rescue with wild-type XPB. In agreement, the XP-B/CS mutation increases KAT2A HAT activity in vitro. Our results unveil a tight connection between TFIIH and KAT2A that controls higher-order chromatin structure and gene expression and provide new insights into transcriptional misregulation in a cancer-prone DNA repair-deficient disorder.


Subject(s)
Chromatin/chemistry , Cockayne Syndrome/genetics , Histone Acetyltransferases/genetics , Histones/metabolism , Protein Subunits/genetics , Transcription Factor TFIIH/genetics , Xeroderma Pigmentosum/genetics , Acetylation , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Chromatin/metabolism , Cockayne Syndrome/metabolism , Cockayne Syndrome/pathology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Editing , Gene Expression Regulation , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Histones/genetics , Humans , Models, Biological , Osteoblasts/cytology , Osteoblasts/metabolism , Primary Cell Culture , Protein Subunits/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factor TFIIH/metabolism , Transcription Initiation, Genetic , Xeroderma Pigmentosum/metabolism , Xeroderma Pigmentosum/pathology
3.
PLoS Genet ; 10(10): e1004732, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25340339

ABSTRACT

The expression of protein-coding genes requires the selective role of many transcription factors, whose coordinated actions remain poorly understood. To further grasp the molecular mechanisms that govern transcription, we focused our attention on the general transcription factor TFIIH, which gives rise, once mutated, to Trichothiodystrophy (TTD), a rare autosomal premature-ageing disease causing inter alia, metabolic dysfunctions. Since this syndrome could be connected to transcriptional defects, we investigated the ability of a TTD mouse model to cope with food deprivation, knowing that energy homeostasis during fasting involves an accurate regulation of the gluconeogenic genes in the liver. Abnormal amounts of gluconeogenic enzymes were thus observed in TTD hepatic parenchyma, which was related to the dysregulation of the corresponding genes. Strikingly, such gene expression defects resulted from the inability of PGC1-α to fulfill its role of coactivator. Indeed, extensive molecular analyses unveiled that wild-type TFIIH cooperated in an ATP-dependent manner with PGC1-α as well as with the deacetylase SIRT1, thereby contributing to the PGC1-α deacetylation by SIRT1. Such dynamic partnership was, however, impaired when TFIIH was mutated, having as a consequence the disruption of PGC1-α recruitment to the promoter of target genes. Therefore, besides a better understanding of the etiology of TFIIH-related disease, our results shed light on the synergistic relationship that exist between different types of transcription factors, which is necessary to properly regulate the expression of protein coding genes.


Subject(s)
Sirtuin 1/genetics , Transcription Factor TFIIH/genetics , Transcription Factors/genetics , Transcription, Genetic , Trichothiodystrophy Syndromes/genetics , Animals , DNA Repair/genetics , Gene Expression Regulation , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Sirtuin 1/biosynthesis , Transcription Factor TFIIH/biosynthesis , Transcription Factors/biosynthesis , Trichothiodystrophy Syndromes/pathology
4.
J Exp Med ; 206(13): 3031-46, 2009 Dec 21.
Article in English | MEDLINE | ID: mdl-19934020

ABSTRACT

Mutations in the XPD subunit of the DNA repair/transcription factor TFIIH result in the rare recessive genetic disorder xeroderma pigmentosum (XP). Many XP patients are compound heterozygotes with a "causative" XPD point mutation R683W and different second mutant alleles, considered "null alleles." However, there is marked clinical heterogeneity (including presence or absence of skin cancers or neurological degeneration) in these XPD/R683W patients, thus suggesting a contribution of the second allele. Here, we report XP patients carrying XPD/R683W and a second XPD allele either XPD/Q452X, /I455del, or /199insPP. We performed a systematic study of the effect of these XPD mutations on several enzymatic functions of TFIIH and found that each mutation exhibited unique biochemical properties. Although all the mutations inhibited the nucleotide excision repair (NER) by disturbing the XPD helicase function, each of them disrupted specific molecular steps during transcription: XPD/Q452X hindered the transactivation process, XPD/I455del disturbed RNA polymerase II phosphorylation, and XPD/199insPP inhibited kinase activity of the cdk7 subunit of TFIIH. The broad range and severity of clinical features in XP patients arise from a broad set of deficiencies in NER and transcription that result from the combination of mutations found on both XPD alleles.


Subject(s)
Xeroderma Pigmentosum Group D Protein/genetics , Xeroderma Pigmentosum/genetics , Adult , Alleles , Animals , Cells, Cultured , Female , Heterozygote , Humans , Kruppel-Like Transcription Factors/analysis , Male , Phenotype , Point Mutation , Spodoptera , Transcription Factor TFIIH/analysis , Transcription, Genetic
5.
Mol Cell ; 16(2): 187-97, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15494306

ABSTRACT

Mutations in the XPD subunit of the transcription/repair factor TFIIH cause the Xeroderma pigmentosum disorder. We show that in some XP-D deficient cells, transactivation by the vitamin D receptor (VDR) is selectively inhibited for a subset of responsive genes, such as CYP24, and that the XPD/R683W mutation prevents VDR recruitment on its promoter. Contrary to other nuclear receptors, VDR, which lacks a functional A/B domain, is not phosphorylated and consequently not regulated by the cdk7 kinase of TFIIH. In fact, we demonstrate that the VDR transactivation defect resides in Ets1, another activator that cannot be phosphorylated by TFIIH in XP-D cells. Indeed, the phosphorylated Ets1 seems to promote the binding of VDR to its responsive element and trigger the subsequent recruitment of coactivators and RNA pol II. We propose a model in which TFIIH regulates the activity of nuclear receptors by phosphorylating either their A/B domain or an additional regulatory DNA binding partner.


Subject(s)
Gene Expression Regulation/physiology , Receptors, Calcitriol/metabolism , Transcription Factors, TFII/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , DNA/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Mutation , Phosphorylation , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Transcription Factor TFIIH , Transcription Factors/genetics , Transcription Factors/metabolism , Vitamin D/metabolism , Vitamin D3 24-Hydroxylase , Xeroderma Pigmentosum Group D Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...