Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell ; 172(3): 439-453.e14, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29290468

ABSTRACT

Telomere maintenance critically depends on the distinct activities of telomerase, which adds telomeric repeats to solve the end replication problem, and RTEL1, which dismantles DNA secondary structures at telomeres to facilitate replisome progression. Here, we establish that reversed replication forks are a pathological substrate for telomerase and the source of telomere catastrophe in Rtel1-/- cells. Inhibiting telomerase recruitment to telomeres, but not its activity, or blocking replication fork reversal through PARP1 inhibition or depleting UBC13 or ZRANB3 prevents the rapid accumulation of dysfunctional telomeres in RTEL1-deficient cells. In this context, we establish that telomerase binding to reversed replication forks inhibits telomere replication, which can be mimicked by preventing replication fork restart through depletion of RECQ1 or PARG. Our results lead us to propose that telomerase inappropriately binds to and inhibits restart of reversed replication forks within telomeres, which compromises replication and leads to critically short telomeres.


Subject(s)
DNA Helicases/genetics , DNA Replication , Telomere Homeostasis , Animals , Cell Line , Cells, Cultured , DNA Helicases/metabolism , Glycoside Hydrolases/metabolism , Mice , Poly (ADP-Ribose) Polymerase-1/metabolism , RecQ Helicases/metabolism , Ubiquitin-Conjugating Enzymes/metabolism
2.
Mol Cell ; 82(10): 1924-1939.e10, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35439434

ABSTRACT

The 53BP1-RIF1-shieldin pathway maintains genome stability by suppressing nucleolytic degradation of DNA ends at double-strand breaks (DSBs). Although RIF1 interacts with damaged chromatin via phospho-53BP1 and facilitates recruitment of the shieldin complex to DSBs, it is unclear whether other regulatory cues contribute to this response. Here, we implicate methylation of histone H3 at lysine 4 by SETD1A-BOD1L in the recruitment of RIF1 to DSBs. Compromising SETD1A or BOD1L expression or deregulating H3K4 methylation allows uncontrolled resection of DNA ends, impairs end-joining of dysfunctional telomeres, and abrogates class switch recombination. Moreover, defects in RIF1 localization to DSBs are evident in patient cells bearing loss-of-function mutations in SETD1A. Loss of SETD1A-dependent RIF1 recruitment in BRCA1-deficient cells restores homologous recombination and leads to resistance to poly(ADP-ribose)polymerase inhibition, reinforcing the clinical relevance of these observations. Mechanistically, RIF1 binds directly to methylated H3K4, facilitating its recruitment to, or stabilization at, DSBs.


Subject(s)
DNA Breaks, Double-Stranded , Telomere-Binding Proteins , BRCA1 Protein/genetics , DNA/metabolism , DNA End-Joining Repair , DNA Repair , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Methylation , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Tumor Suppressor p53-Binding Protein 1/metabolism
3.
Mol Cell ; 81(4): 767-783.e11, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33333017

ABSTRACT

Chromatin is a barrier to efficient DNA repair, as it hinders access and processing of certain DNA lesions. ALC1/CHD1L is a nucleosome-remodeling enzyme that responds to DNA damage, but its precise function in DNA repair remains unknown. Here we report that loss of ALC1 confers sensitivity to PARP inhibitors, methyl-methanesulfonate, and uracil misincorporation, which reflects the need to remodel nucleosomes following base excision by DNA glycosylases but prior to handover to APEX1. Using CRISPR screens, we establish that ALC1 loss is synthetic lethal with homologous recombination deficiency (HRD), which we attribute to chromosome instability caused by unrepaired DNA gaps at replication forks. In the absence of ALC1 or APEX1, incomplete processing of BER intermediates results in post-replicative DNA gaps and a critical dependence on HR for repair. Hence, targeting ALC1 alone or as a PARP inhibitor sensitizer could be employed to augment existing therapeutic strategies for HRD cancers.


Subject(s)
Chromatin Assembly and Disassembly , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Nucleosomes/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , DNA Helicases/genetics , DNA Replication/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-Binding Proteins/genetics , Homologous Recombination/drug effects , Mice , Mice, Knockout , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Nucleosomes/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics
4.
Nature ; 589(7840): 103-109, 2021 01.
Article in English | MEDLINE | ID: mdl-33239783

ABSTRACT

Mammalian telomeres protect chromosome ends from aberrant DNA repair1. TRF2, a component of the telomere-specific shelterin protein complex, facilitates end protection through sequestration of the terminal telomere repeat sequence within a lariat T-loop structure2,3. Deleting TRF2 (also known as TERF2) in somatic cells abolishes T-loop formation, which coincides with telomere deprotection, chromosome end-to-end fusions and inviability3-9. Here we establish that, by contrast, TRF2 is largely dispensable for telomere protection in mouse pluripotent embryonic stem (ES) and epiblast stem cells. ES cell telomeres devoid of TRF2 instead activate an attenuated telomeric DNA damage response that lacks accompanying telomere fusions, and propagate for multiple generations. The induction of telomere dysfunction in ES cells, consistent with somatic deletion of Trf2 (also known as Terf2), occurs only following the removal of the entire shelterin complex. Consistent with TRF2 being largely dispensable for telomere protection specifically during early embryonic development, cells exiting pluripotency rapidly switch to TRF2-dependent end protection. In addition, Trf2-null embryos arrest before implantation, with evidence of strong DNA damage response signalling and apoptosis specifically in the non-pluripotent compartment. Finally, we show that ES cells form T-loops independently of TRF2, which reveals why TRF2 is dispensable for end protection during pluripotency. Collectively, these data establish that telomere protection is solved by distinct mechanisms in pluripotent and somatic tissues.


Subject(s)
Chromosomes, Mammalian/metabolism , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/deficiency , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Survival , Chromosomes, Mammalian/genetics , Germ Layers/cytology , Germ Layers/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Telomere/genetics , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism
5.
Mol Cell ; 70(4): 707-721.e7, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29754823

ABSTRACT

DNA polymerase ε (POLE) is a four-subunit complex and the major leading strand polymerase in eukaryotes. Budding yeast orthologs of POLE3 and POLE4 promote Polε processivity in vitro but are dispensable for viability in vivo. Here, we report that POLE4 deficiency in mice destabilizes the entire Polε complex, leading to embryonic lethality in inbred strains and extensive developmental abnormalities, leukopenia, and tumor predisposition in outbred strains. Comparable phenotypes of growth retardation and immunodeficiency are also observed in human patients harboring destabilizing mutations in POLE1. In both Pole4-/- mouse and POLE1 mutant human cells, Polε hypomorphy is associated with replication stress and p53 activation, which we attribute to inefficient replication origin firing. Strikingly, removing p53 is sufficient to rescue embryonic lethality and all developmental abnormalities in Pole4 null mice. However, Pole4-/-p53+/- mice exhibit accelerated tumorigenesis, revealing an important role for controlled CMG and origin activation in normal development and tumor prevention.


Subject(s)
Carcinogenesis/pathology , DNA Polymerase II/chemistry , DNA Polymerase II/physiology , DNA Replication , Developmental Disabilities/etiology , Growth Disorders/etiology , Leukopenia/etiology , Animals , Carcinogenesis/genetics , Cells, Cultured , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Humans , Infant, Newborn , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Tumor Suppressor Protein p53/physiology
6.
Nature ; 575(7783): 523-527, 2019 11.
Article in English | MEDLINE | ID: mdl-31723267

ABSTRACT

The protection of telomere ends by the shelterin complex prevents DNA damage signalling and promiscuous repair at chromosome ends. Evidence suggests that the 3' single-stranded telomere end can assemble into a lasso-like t-loop configuration1,2, which has been proposed to safeguard chromosome ends from being recognized as DNA double-strand breaks2. Mechanisms must also exist to transiently disassemble t-loops to allow accurate telomere replication and to permit telomerase access to the 3' end to solve the end-replication problem. However, the regulation and physiological importance of t-loops in the protection of telomere ends remains unknown. Here we identify a CDK phosphorylation site in the shelterin subunit at Ser365 of TRF2, whose dephosphorylation in S phase by the PP6R3 phosphatase provides a narrow window during which the RTEL1 helicase can transiently access and unwind t-loops to facilitate telomere replication. Re-phosphorylation of TRF2 at Ser365 outside of S phase is required to release RTEL1 from telomeres, which not only protects t-loops from promiscuous unwinding and inappropriate activation of ATM, but also counteracts replication conflicts at DNA secondary structures that arise within telomeres and across the genome. Hence, a phospho-switch in TRF2 coordinates the assembly and disassembly of t-loops during the cell cycle, which protects telomeres from replication stress and an unscheduled DNA damage response.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinases/metabolism , Phosphoserine/metabolism , Telomere/metabolism , Telomeric Repeat Binding Protein 2/chemistry , Telomeric Repeat Binding Protein 2/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA/biosynthesis , DNA/chemistry , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Damage , DNA Helicases/metabolism , DNA Repair , DNA Replication , Fibroblasts , Genome/genetics , HEK293 Cells , Humans , Mice , Mutation , Phenotype , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , S Phase , Shelterin Complex , Telomerase/metabolism , Telomere/genetics , Telomere-Binding Proteins/chemistry , Telomere-Binding Proteins/metabolism , Telomeric Repeat Binding Protein 2/genetics
8.
Mol Cell ; 59(3): 462-77, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26166705

ABSTRACT

Recognition and repair of damaged replication forks are essential to maintain genome stability and are coordinated by the combined action of the Fanconi anemia and homologous recombination pathways. These pathways are vital to protect stalled replication forks from uncontrolled nucleolytic activity, which otherwise causes irreparable genomic damage. Here, we identify BOD1L as a component of this fork protection pathway, which safeguards genome stability after replication stress. Loss of BOD1L confers exquisite cellular sensitivity to replication stress and uncontrolled resection of damaged replication forks, due to a failure to stabilize RAD51 at these forks. Blocking DNA2-dependent resection, or downregulation of the helicases BLM and FBH1, suppresses both catastrophic fork processing and the accumulation of chromosomal damage in BOD1L-deficient cells. Thus, our work implicates BOD1L as a critical regulator of genome integrity that restrains nucleolytic degradation of damaged replication forks.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Replication , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Cell Line , Cell Survival , DNA Damage , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Genome, Human , Genomic Instability , HeLa Cells , Humans , RecQ Helicases/metabolism
9.
Genes Dev ; 29(24): 2532-46, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26637282

ABSTRACT

Microsatellites are short tandem repeat sequences that are highly prone to expansion/contraction due to their propensity to form non-B-form DNA structures, which hinder DNA polymerases and provoke template slippage. Although error correction by mismatch repair plays a key role in preventing microsatellite instability (MSI), which is a hallmark of Lynch syndrome, activities must also exist that unwind secondary structures to facilitate replication fidelity. Here, we report that Fancj helicase-deficient mice, while phenotypically resembling Fanconi anemia (FA), are also hypersensitive to replication inhibitors and predisposed to lymphoma. Whereas metabolism of G4-DNA structures is largely unaffected in Fancj(-/-) mice, high levels of spontaneous MSI occur, which is exacerbated by replication inhibition. In contrast, MSI is not observed in Fancd2(-/-) mice but is prevalent in human FA-J patients. Together, these data implicate FANCJ as a key factor required to counteract MSI, which is functionally distinct from its role in the FA pathway.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia/physiopathology , Lymphoma/genetics , Microsatellite Instability , Animals , Antineoplastic Agents/pharmacology , Basic-Leucine Zipper Transcription Factors/deficiency , Camptothecin/pharmacology , Cell Line , Cells, Cultured , DNA Damage/genetics , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group Proteins/deficiency , Female , Fibroblasts/drug effects , Fibroblasts/radiation effects , Genetic Predisposition to Disease , Humans , Male , Mice, Knockout , Mitomycin/pharmacology , Neoplasms, Glandular and Epithelial/genetics , RNA Helicases , Ultraviolet Rays
10.
Am J Hum Genet ; 103(6): 1038-1044, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30503519

ABSTRACT

During genome replication, polymerase epsilon (Pol ε) acts as the major leading-strand DNA polymerase. Here we report the identification of biallelic mutations in POLE, encoding the Pol ε catalytic subunit POLE1, in 15 individuals from 12 families. Phenotypically, these individuals had clinical features closely resembling IMAGe syndrome (intrauterine growth restriction [IUGR], metaphyseal dysplasia, adrenal hypoplasia congenita, and genitourinary anomalies in males), a disorder previously associated with gain-of-function mutations in CDKN1C. POLE1-deficient individuals also exhibited distinctive facial features and variable immune dysfunction with evidence of lymphocyte deficiency. All subjects shared the same intronic variant (c.1686+32C>G) as part of a common haplotype, in combination with different loss-of-function variants in trans. The intronic variant alters splicing, and together the biallelic mutations lead to cellular deficiency of Pol ε and delayed S-phase progression. In summary, we establish POLE as a second gene in which mutations cause IMAGe syndrome. These findings add to a growing list of disorders due to mutations in DNA replication genes that manifest growth restriction alongside adrenal dysfunction and/or immunodeficiency, consolidating these as replisome phenotypes and highlighting a need for future studies to understand the tissue-specific development roles of the encoded proteins.


Subject(s)
Adrenal Insufficiency/genetics , DNA Polymerase II/genetics , Fetal Growth Retardation/genetics , Mutation/genetics , Osteochondrodysplasias/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Urogenital Abnormalities/genetics , Adolescent , Adult , Alleles , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p57/genetics , DNA Replication/genetics , Female , Humans , Infant , Male , Middle Aged , Phenotype , Young Adult
11.
Mol Cell ; 49(5): 858-71, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23333305

ABSTRACT

The appropriate execution of DNA double-strand break (DSB) repair is critical for genome stability and tumor avoidance. 53BP1 and BRCA1 directly influence DSB repair pathway choice by regulating 5' end resection, but how this is achieved remains uncertain. Here we report that Rif1(-/-) mice are severely compromised for 53BP1-dependent class switch recombination (CSR) and fusion of dysfunctional telomeres. The inappropriate accumulation of RIF1 at DSBs in S phase is antagonized by BRCA1, and deletion of Rif1 suppresses toxic nonhomologous end joining (NHEJ) induced by PARP inhibition in Brca1-deficient cells. Mechanistically, RIF1 is recruited to DSBs via the N-terminal phospho-SQ/TQ domain of 53BP1, and DSBs generated by ionizing radiation or during CSR are hyperresected in the absence of RIF1. Thus, RIF1 and 53BP1 cooperate to block DSB resection to promote NHEJ in G1, which is antagonized by BRCA1 in S phase to ensure a switch of DSB repair mode to homologous recombination.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Binding Proteins/genetics , DNA/metabolism , Telomere-Binding Proteins/genetics , Animals , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Mice , Recombination, Genetic , Telomere/metabolism , Telomere-Binding Proteins/metabolism , Transfection , Tumor Suppressor p53-Binding Protein 1
12.
Nature ; 502(7471): 381-4, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24005329

ABSTRACT

Repair of interstrand crosslinks (ICLs) requires the coordinated action of the intra-S-phase checkpoint and the Fanconi anaemia pathway, which promote ICL incision, translesion synthesis and homologous recombination (reviewed in refs 1, 2). Previous studies have implicated the 3'-5' superfamily 2 helicase HELQ in ICL repair in Drosophila melanogaster (MUS301 (ref. 3)) and Caenorhabditis elegans (HELQ-1 (ref. 4)). Although in vitro analysis suggests that HELQ preferentially unwinds synthetic replication fork substrates with 3' single-stranded DNA overhangs and also disrupts protein-DNA interactions while translocating along DNA, little is known regarding its functions in mammalian organisms. Here we report that HELQ helicase-deficient mice exhibit subfertility, germ cell attrition, ICL sensitivity and tumour predisposition, with Helq heterozygous mice exhibiting a similar, albeit less severe, phenotype than the null, indicative of haploinsufficiency. We establish that HELQ interacts directly with the RAD51 paralogue complex BCDX2 and functions in parallel to the Fanconi anaemia pathway to promote efficient homologous recombination at damaged replication forks. Thus, our results reveal a critical role for HELQ in replication-coupled DNA repair, germ cell maintenance and tumour suppression in mammals.


Subject(s)
Carcinogenesis , DNA Helicases/metabolism , DNA Repair , Germ Cells/metabolism , Germ Cells/pathology , Rad51 Recombinase/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , DNA Damage/genetics , DNA Helicases/deficiency , DNA Helicases/genetics , DNA Repair/genetics , DNA Replication/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Female , Gene Deletion , Germ Cells/cytology , Male , Mice , Multiprotein Complexes/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovary/metabolism , Ovary/pathology , Recombinational DNA Repair/genetics
13.
Biol Reprod ; 86(2): 48, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22053096

ABSTRACT

It has been proposed that four members of the aquaporin family (AQPs 1, 3, 8, and 9) are involved in the control of amniotic fluid (AF) homeostasis, as illustrated by their differential expression patterns in normal and pathological human term fetal membranes. However, there are no data available to date on their ontogeny throughout pregnancy. Our objective was to determine spatiotemporal expression profiles of the mRNA and proteins of all 13 members of this transmembrane channel family. For this purpose, we used healthy fetal membranes from the first, second, and third trimesters of pregnancy. Total mRNA and proteins were extracted from total membranes and from separated amnion and chorion. Quantitative PCR, Western blot, and immunohistochemistry experiments were carried out to determine the presence of AQPs and to quantify their spatiotemporal expression patterns throughout pregnancy. The WISH cell line was tested to propose a cellular model for the role of AQPs in the amnion compartment. AQP11 expression was established in amniotic membranes at term. Aquaporins 1, 3, 8, 9, and 11 mRNA and proteins were present in amnion and chorion throughout human gestation. Each AQP has a time-specific expression pattern, with AQP1 presenting the highest variation in terms of mRNA and protein levels. The WISH cell line also expressed the same five AQPs. Taken together, these results indicate that AQPs are expressed and potentially involved in the regulation of AF homeostasis throughout pregnancy. This also clearly supports the hypothesis that abnormal expression could occur at any time during pregnancy, ultimately leading to obstetrical pathologies such as polyhydramnios or oligohydramnios.


Subject(s)
Amnion/metabolism , Aquaporins/metabolism , Chorion/metabolism , Extraembryonic Membranes/metabolism , Pregnancy Trimester, First/metabolism , Pregnancy Trimester, Second/metabolism , Pregnancy Trimester, Third/metabolism , Adult , Amnion/cytology , Amniotic Fluid/metabolism , Aquaporin 1/metabolism , Aquaporin 3/metabolism , Cell Line , Female , Homeostasis , Humans , Pregnancy , RNA, Messenger/metabolism
14.
Cell Rep ; 39(9): 110871, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35649380

ABSTRACT

The maintenance of genome stability relies on coordinated control of origin activation and replication fork progression. How the interplay between these processes influences human genetic disease and cancer remains incompletely characterized. Here we show that mouse cells featuring Polε instability exhibit impaired genome-wide activation of DNA replication origins, in an origin-location-independent manner. Strikingly, Trp53 ablation in primary Polε hypomorphic cells increased Polε levels and origin activation and reduced DNA damage in a transcription-dependent manner. Transcriptome analysis of primary Trp53 knockout cells revealed that the TRP53-CDKN1A/P21 axis maintains appropriate levels of replication factors and CDK activity during unchallenged S phase. Loss of this control mechanism deregulates origin activation and perturbs genome-wide replication fork progression. Thus, while our data support an impaired origin activation model for genetic diseases affecting CMG formation, we propose that loss of the TRP53-CDKN1A/P21 tumor suppressor axis induces inappropriate origin activation and deregulates genome-wide fork progression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21 , DNA Polymerase II , DNA Replication , Poly-ADP-Ribose Binding Proteins , Replication Origin , Tumor Suppressor Protein p53 , Animals , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Damage/genetics , DNA Polymerase II/genetics , DNA Replication/genetics , Mice , Poly-ADP-Ribose Binding Proteins/genetics , S Phase , Tumor Suppressor Protein p53/genetics
15.
J Cell Mol Med ; 14(6B): 1793-805, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19538480

ABSTRACT

The collagenolytic effects of the tissue-type plasminogen activator (t-PA) leading to extracellular matrix degradation are clearly involved in the physiopathology of human foetal membranes rupture. Nevertheless, the regulation of t-PA gene expression in extraembryonic developmental contexts remains unknown. The aim of our study is to propose the retinoic acids (RAs) as molecular regulators of t-PA expression in foetal membranes. RA induced t-PA mRNA and proteins in a time-dependent manner in amniotic membrane explants and Wistar Institute Susan Hayflick (WISH) cells. Furthermore, the use of cycloheximide revealed a two-step regulation of t-PA gene. Gene reporter assays confirmed that the RA-induced t-PA gene expression occurred through interactions of retinoid receptors (RARs and RXRs) with a DR5 response element located at -7 kb from the transcription site. Site-directed mutagenesis of this region of the t-PA promoter showed that SP1 factor was also retinoid-mediated induction, and immunoprecipitation assays revealed that SP1 and RAR/RXR interacted physically. Chromatin immunoprecipitation demonstrated that interactions between RARs, RXRs and t-PA promoter were time dependent: RAR-alpha/RXR-alpha bound DR5 motif before and up to 12 hrs of RA exposure, and RAR-beta/RXR-alpha bound DR5 response element after 12 hrs of RA treatment. Finally, experiments using shRNA and RAR-beta-specific antagonist revealed that reducing RAR-beta induction decreased t-PA induction. Altogether, our results established that the RA-mediated regulation of t-PA in human foetal membranes occurred through two steps, with a major role played by RAR-beta.


Subject(s)
Amnion/drug effects , Amnion/metabolism , Gene Expression Regulation/drug effects , Retinoids/pharmacology , Tissue Plasminogen Activator/genetics , Cell Line , Dibenzazepines/pharmacology , Humans , Protein Binding/drug effects , RNA, Small Interfering/metabolism , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Response Elements/genetics , Retinoid X Receptors/metabolism , Sp1 Transcription Factor/metabolism , Tissue Plasminogen Activator/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
16.
Cell Rep ; 31(8): 107675, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32460026

ABSTRACT

Genome stability requires coordination of DNA replication origin activation and replication fork progression. RTEL1 is a regulator of homologous recombination (HR) implicated in meiotic cross-over control and DNA repair in C. elegans. Through a genome-wide synthetic lethal screen, we uncovered an essential genetic interaction between RTEL1 and DNA polymerase (Pol) epsilon. Loss of POLE4, an accessory subunit of Pol epsilon, has no overt phenotype in worms. In contrast, the combined loss of POLE-4 and RTEL-1 results in embryonic lethality, accumulation of HR intermediates, genome instability, and cessation of DNA replication. Similarly, loss of Rtel1 in Pole4-/- mouse cells inhibits cellular proliferation, which is associated with persistent HR intermediates and incomplete DNA replication. We propose that RTEL1 facilitates genome-wide fork progression through its ability to metabolize DNA secondary structures that form during DNA replication. Loss of this function becomes incompatible with cell survival under conditions of reduced origin activation, such as Pol epsilon hypomorphy.


Subject(s)
DNA Helicases/genetics , DNA Polymerase II/genetics , DNA Replication/genetics , Genomic Instability/genetics , Animals , Humans
17.
Science ; 337(6091): 243-5, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22678361

ABSTRACT

The function of many DNA metabolism proteins depends on their ability to coordinate an iron-sulfur (Fe-S) cluster. Biogenesis of Fe-S proteins is a multistep process that takes place in mitochondria and the cytoplasm, but how it is linked to nuclear Fe-S proteins is not known. Here, we demonstrate that MMS19 forms a complex with the cytoplasmic Fe-S assembly (CIA) proteins CIAO1, IOP1, and MIP18. Cytoplasmic MMS19 also binds to multiple nuclear Fe-S proteins involved in DNA metabolism. In the absence of MMS19, a failure to transfer Fe-S clusters to target proteins is associated with Fe-S protein instability and preimplantation death of mice in which Mms19 has been knocked out. We propose that MMS19 functions as a platform to facilitate Fe-S cluster transfer to proteins critical for DNA replication and repair.


Subject(s)
Cytoplasm/metabolism , DNA/metabolism , Iron-Sulfur Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , DNA Repair , DNA Replication , Humans , Hydrogenase/metabolism , Metallochaperones/metabolism , Metalloproteins , Mice , Mice, Knockout , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Stability , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcription Factors/genetics , Xeroderma Pigmentosum Group D Protein/metabolism
18.
PPAR Res ; 2008: 758562, 2008.
Article in English | MEDLINE | ID: mdl-18288292

ABSTRACT

The placenta is a transitory structure indispensable for the proper development of the embryo and fetus during mammalian gestation. Like other members of the nuclear receptor family, the peroxisome proliferator-activated receptors (PPARs) are known to be involved in the physiological and pathological events occurring during the placentation. This placental involvement has been recently reviewed focusing on the early stages of placental development (implantation and invasion, etc.), mouse PPARs knockout phenotypes, and cytotrophoblast physiology. In this review, we describe the placental involvement of PPARs (e.g., fat transport and metabolism, etc.) during the late stages of gestation and in the amniotic membranes, highlighting their roles in the inflammation process (e.g., chorioamnionitis), metabolic disorders (e.g., diabetes), and parturition.

19.
Biochem Biophys Res Commun ; 346(4): 1207-16, 2006 Aug 11.
Article in English | MEDLINE | ID: mdl-16793012

ABSTRACT

Vitamin A (retinol) and its active derivatives (the retinoids) are essential for the growth and development of the mammalian fetus and placenta. The amniotic membranes are extra-embryonic structures that are indispensable for normal gestation in mammals. Although placental involvement of retinoids is clearly established, little is known about the roles of retinoids for the associated amniotic membranes. The aim of this study was to define the metabolic and molecular pathways of retinoic signaling in human fetal membranes. The expression of retinoid receptors (RARalpha, beta and RXRalpha, beta) was established at transcript and protein levels. Enzymes involved in retinoic acid generation were also detected. The enzymatic generation of functional retinoids was confirmed using specific inhibitors of retinol metabolism. Finally, the functionality of retinoid pathways was demonstrated by inducing established retinoid target gene expression. Our results clearly demonstrated that the molecular and metabolic actors of retinoic signaling pathways are functional in human fetal membranes.


Subject(s)
Amnion/metabolism , Retinoids/metabolism , Female , Humans , Pregnancy , Tretinoin/metabolism , Vitamin A/metabolism
20.
Biochem Biophys Res Commun ; 343(3): 745-53, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16563348

ABSTRACT

Pregnancy-specific glycoproteins (PSGs) are major placental proteins thought to be essential for the maintenance of gestation. Little is known about the regulation of expression of the 11 genes encoding these proteins. It was previously demonstrated that Krüppel-like factor 6 (KLF6) and specific-protein 1 (Sp1) bind to conserved sequence within the PSG-5 gene promoter. Informatics analysis revealed the presence of one potential binding site for Krüppel-like factor 4 (KLF4), in the PSG-5 promoter, suggesting a potential transcriptional regulator role for KLF4. Using gene promoter-reporter transfections and X-ChIP assays, we demonstrated that KLF4 is an activator of the PSG-5 promoter by binding to a KLF consensus like binding which includes the Core Promoter Element region (-147/-140). Furthermore, we used previous data showing the binding of Sp1 transcription factor to a GT-box (-443/-437) and co-transfection assays with KLF4 and Sp1 to demonstrate the strong synergic activity of these two factors on the PSG-5 promoter.


Subject(s)
Glycoproteins/genetics , Kruppel-Like Transcription Factors/metabolism , Pregnancy-Specific beta 1-Glycoproteins/genetics , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Transcriptional Activation , Binding Sites , Cell Line , HeLa Cells , Humans , Kruppel-Like Factor 4
SELECTION OF CITATIONS
SEARCH DETAIL