Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Colloids Surf B Biointerfaces ; 224: 113219, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36848782

ABSTRACT

The design and preparation of new vectors to transport genetic material and increase the transfection efficiency continue being an important research line. Here, a novel biocompatible sugar-based polymer derived from D-mannitol has been synthesized to be used as a gene material nanocarrier in human (gene transfection) and microalga cells (transformation process). Its low toxicity allows its use in processes with both medical and industrial applications. A multidisciplinary study about the formation of polymer/p-DNA polyplexes has been carried out using techniques such as gel electrophoresis, zeta potential, dynamic light scattering, atomic force microscopy, and circular dichroism spectroscopy. The nucleic acids used were the eukaryotic expression plasmid pEGFP-C1 and the microalgal expression plasmid Phyco69, which showed different behaviors. The importance of DNA supercoiling in both transfection and transformation processes was demonstrated. Better results were obtained in microalga cells nuclear transformation than in human cells gene transfection. This was related to the plasmid's conformational changes, in particular to their superhelical structure. It is noteworthy that the same nanocarrier has been used with eukaryotic cells from both human and microalga.


Subject(s)
Eukaryotic Cells , Polymers , Humans , Polymers/chemistry , Mannitol , Transfection , Plasmids/genetics , DNA/chemistry , Genetic Engineering , Genetic Vectors/genetics
2.
Cell Death Dis ; 13(5): 503, 2022 05 27.
Article in English | MEDLINE | ID: mdl-35624090

ABSTRACT

Fanconi anemia (FA) is a rare hereditary disorder caused by mutations in any one of the FANC genes. FA cells are mainly characterized by extreme hypersensitivity to interstrand crosslink (ICL) agents. Additionally, the FA proteins play a crucial role in concert with homologous recombination (HR) factors to protect stalled replication forks. Here, we report that the 5-methyl-2'-deoxycytidine (5mdC) demethylation (pathway) intermediate 5-hydroxymethyl-2'-deoxycytidine (5hmdC) and its deamination product 5-hydroxymethyl-2'-deoxyuridine (5hmdU) elicit a DNA damage response, chromosome aberrations, replication fork impairment and cell viability loss in the absence of FANCD2. Interestingly, replication fork instability by 5hmdC or 5hmdU was associated to the presence of Poly(ADP-ribose) polymerase 1 (PARP1) on chromatin, being both phenotypes exacerbated by olaparib treatment. Remarkably, Parp1-/- cells did not show any replication fork defects or sensitivity to 5hmdC or 5hmdU, suggesting that retained PARP1 at base excision repair (BER) intermediates accounts for the observed replication fork defects upon 5hmdC or 5hmdU incorporation in the absence of FANCD2. We therefore conclude that 5hmdC is deaminated in vivo to 5hmdU, whose fixation by PARP1 during BER, hinders replication fork progression and contributes to genomic instability in FA cells.


Subject(s)
Fanconi Anemia , DNA Demethylation , DNA Replication , Deoxycytidine/analogs & derivatives , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Humans , Thymidine/analogs & derivatives
3.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35055077

ABSTRACT

Whilst avoidance of chemical modifications of DNA bases is essential to maintain genome stability, during evolution eukaryotic cells have evolved a chemically reversible modification of the cytosine base. These dynamic methylation and demethylation reactions on carbon-5 of cytosine regulate several cellular and developmental processes such as embryonic stem cell pluripotency, cell identity, differentiation or tumourgenesis. Whereas these physiological processes are well characterized, very little is known about the toxicity of these cytosine analogues when they incorporate during replication. Here, we report a role of the base excision repair factor XRCC1 in protecting replication fork upon incorporation of 5-hydroxymethyl-2'-deoxycytosine (5hmC) and its deamination product 5-hydroxymethyl-2'-deoxyuridine (5hmU) during DNA synthesis. In the absence of XRCC1, 5hmC exposure leads to increased genomic instability, replication fork impairment and cell lethality. Moreover, the 5hmC deamination product 5hmU recapitulated the genomic instability phenotypes observed by 5hmC exposure, suggesting that 5hmU accounts for the observed by 5hmC exposure. Remarkably, 5hmC-dependent genomic instability and replication fork impairment seen in Xrcc1-/- cells were exacerbated by the trapping of Parp1 on chromatin, indicating that XRCC1 maintains replication fork stability during processing of 5hmC and 5hmU by the base excision repair pathway. Our findings uncover natural epigenetic DNA bases 5hmC and 5hmU as genotoxic nucleosides that threaten replication dynamics and genome integrity in the absence of XRCC1.


Subject(s)
DNA Demethylation , DNA Replication , Deoxycytidine/analogs & derivatives , Thymidine/analogs & derivatives , X-ray Repair Cross Complementing Protein 1/genetics , 5-Methylcytosine/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , DNA Damage , DNA Replication/drug effects , Epigenesis, Genetic , Genomic Instability , Humans , Replication Origin , X-ray Repair Cross Complementing Protein 1/metabolism
4.
Nucleic Acids Res ; 49(1): 206-220, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33330942

ABSTRACT

Proteostasis needs to be tightly controlled to meet the cellular demand for correctly de novo folded proteins and to avoid protein aggregation. While a coupling between translation rate and co-translational folding, likely involving an interplay between the ribosome and its associated chaperones, clearly appears to exist, the underlying mechanisms and the contribution of ribosomal proteins remain to be explored. The ribosomal protein uL3 contains a long internal loop whose tip region is in close proximity to the ribosomal peptidyl transferase center. Intriguingly, the rpl3[W255C] allele, in which the residue making the closest contact to this catalytic site is mutated, affects diverse aspects of ribosome biogenesis and function. Here, we have uncovered, by performing a synthetic lethal screen with this allele, an unexpected link between translation and the folding of nascent proteins by the ribosome-associated Ssb-RAC chaperone system. Our results reveal that uL3 and Ssb-RAC cooperate to prevent 80S ribosomes from piling up within the 5' region of mRNAs early on during translation elongation. Together, our study provides compelling in vivo evidence for a functional connection between peptide bond formation at the peptidyl transferase center and chaperone-assisted de novo folding of nascent polypeptides at the solvent-side of the peptide exit tunnel.


Subject(s)
Molecular Chaperones/physiology , Multiprotein Complexes/physiology , Peptide Chain Elongation, Translational/physiology , Protein Folding , Proteostasis/physiology , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/metabolism , Alleles , Loss of Function Mutation , Molecular Chaperones/genetics , Mutation, Missense , Peptidyl Transferases/physiology , Point Mutation , Recombinant Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/physiology , Ribosomes/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
5.
Pharmaceutics ; 12(5)2020 May 25.
Article in English | MEDLINE | ID: mdl-32466339

ABSTRACT

Gene therapy is a therapeutic process consisting of the transport of genetic material into cells. The design and preparation of novel carriers to transport DNA is an important research line in the medical field. Hybrid compounds such as metallo-liposomes, containing a mixture of lipids, were prepared and characterized. Cationic metal lipids derived from the [Ru(bpy)3]2+ complex, RuC11C11 or RuC19C19, both with different hydrophobic/lipophilic ratios, were mixed with the phospholipid DOPE. A relation between the size and the molar fraction α was found and a multidisciplinary study about the interaction between the metallo-liposomes and DNA was performed. The metallo-liposomes/DNA association was quantified and a relationship between Kapp and α was obtained. Techniques such as AFM, SEM, zeta potential, dynamic light scattering and agarose gel electrophoresis demonstrated the formation of lipoplexes and showed the structure of the liposomes. L/D values corresponding to the polynucleotide's condensation were estimated. In vitro assays proved the low cell toxicity of the metallo-liposomes, lower for normal cells than for cancer cell lines, and a good internalization into cells. The latter as well as the transfection measurements carried out with plasmid DNA pEGFP-C1 have demonstrated a good availability of the Ru(II)-based liposomes for being used as non-toxic nanovectors in gene therapy.

6.
Cell Death Differ ; 25(11): 1967-1979, 2018 11.
Article in English | MEDLINE | ID: mdl-29511342

ABSTRACT

Metabolically reactive formaldehyde is a genotoxin and a carcinogen. Mice lacking the main formaldehyde-detoxifying gene Adh5 combined with the loss of the Fanconi anemia (FA) DNA repair pathway rapidly succumbed to bone marrow failure (BMF) primarily due to the extensive ablation of the hematopoietic stem cell (HSC) pool. However, the mechanism by which formaldehyde mediates these toxic effects is still unknown. We uncover a detrimental role of tetrahydrofolic acid (THF) in cells lacking Adh5 or the FA repair pathway. We show that Adh5- or FA-deficient cells are hypersensitive to formaldehyde and to THF, presenting DNA damage and genome instability. THF cytotoxicity involved imbalance of the nucleotide pool by deregulation of the thymidylate synthase (TYMS) enzyme, which stalled replication forks. In mice, THF exposure had widespread effects on hematopoiesis, affecting the frequency and the viability of myeloid- and lymphoid-committed precursor cells. Moreover, the hematopoietic stem and progenitor cells (HSPC) showed genomic instability, reduced colony-forming capacity and increased frequency of cycling and apoptotic HSCs upon THF exposure. Overall, our data reveal that the physiological pool of THF and formaldehyde challenge the stability of the genome of HSPCs that might lead to blood disorders.


Subject(s)
DNA Damage/drug effects , DNA Repair/drug effects , Tetrahydrofolates/toxicity , Alcohol Dehydrogenase/deficiency , Alcohol Dehydrogenase/genetics , Animals , Apoptosis/drug effects , Cell Line , Chickens , Fanconi Anemia Complementation Group Proteins/deficiency , Fanconi Anemia Complementation Group Proteins/genetics , Genomic Instability/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Histones/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Thymidylate Synthase/metabolism
7.
Nature ; 548(7669): 549-554, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28813411

ABSTRACT

The folate-driven one-carbon (1C) cycle is a fundamental metabolic hub in cells that enables the synthesis of nucleotides and amino acids and epigenetic modifications. This cycle might also release formaldehyde, a potent protein and DNA crosslinking agent that organisms produce in substantial quantities. Here we show that supplementation with tetrahydrofolate, the essential cofactor of this cycle, and other oxidation-prone folate derivatives kills human, mouse and chicken cells that cannot detoxify formaldehyde or that lack DNA crosslink repair. Notably, formaldehyde is generated from oxidative decomposition of the folate backbone. Furthermore, we find that formaldehyde detoxification in human cells generates formate, and thereby promotes nucleotide synthesis. This supply of 1C units is sufficient to sustain the growth of cells that are unable to use serine, which is the predominant source of 1C units. These findings identify an unexpected source of formaldehyde and, more generally, indicate that the detoxification of this ubiquitous endogenous genotoxin creates a benign 1C unit that can sustain essential metabolism.


Subject(s)
Carbon/metabolism , Folic Acid/chemistry , Folic Acid/metabolism , Formaldehyde/chemistry , Formaldehyde/metabolism , Metabolic Networks and Pathways , Mutagens/chemistry , Mutagens/metabolism , Alcohol Dehydrogenase/metabolism , Animals , Carbon/deficiency , Cell Line , Chickens , Coenzymes/metabolism , Cross-Linking Reagents/metabolism , DNA Damage , DNA Repair , Humans , Inactivation, Metabolic , Mice , Nucleotides/biosynthesis , Oxidation-Reduction , Serine/chemistry , Serine/metabolism , Tetrahydrofolates/metabolism
8.
Chem Asian J ; 12(6): 679-689, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28112869

ABSTRACT

Given the high toxicity of the anthracycline antibiotic doxorubicin (DOX), it is relevant to search for nanocarriers that decrease the side effects of the drug and are able to transport it towards a therapeutic target Here, the encapsulation of DOX by p-sulfocalix[6]arene (calix) has been studied. The interaction of DOX with the macrocycle, as well as with DNA, has been investigated and the equilibrium constant for each binding process estimated. The results showed that the binding constant of DOX to DNA, KDNA , is three orders of magnitude higher than that to calix, Kcalix . The ability of calixarenes to encapsulate DOX molecules, as well as the capability of the DOX molecules included into the inner cavity of the macrocycle to bind with DNA have been examined. Cytotoxicity measurements were done in different cancer and normal cell lines to probe the decrease in the toxicity of the encapsulated DOX. The low toxicity of calixarenes has also been demonstrated for different cell lines.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Calixarenes/chemistry , Doxorubicin/administration & dosage , Drug Delivery Systems , Nanoparticles/chemistry , Phenols/chemistry , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA, Neoplasm/drug effects , Dose-Response Relationship, Drug , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Structure-Activity Relationship
10.
Cell Death Dis ; 7(12): e2516, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27929539

ABSTRACT

It is established that hematopoietic stem cells (HSC) in the hypoxic bone marrow have adapted their metabolism to oxygen-limiting conditions. This adaptation includes suppression of mitochondrial activity, induction of anerobic glycolysis, and activation of hypoxia-inducible transcription factor 1α (Hif1α)-dependent gene expression. During progression of hematopoiesis, a metabolic switch towards mitochondrial oxidative phosphorylation is observed, making this organelle essential for determining cell fate choice in bone marrow. However, given that HSC metabolism is essentially oxygen-independent, it is still unclear whether functional mitochondria are absolutely required for their survival. To assess the actual dependency of these undifferentiated cells on mitochondrial function, we have performed an analysis of the hematopoiesis in a mouse mutant, named SDHD-ESR, with inducible deletion of the mitochondrial protein-encoding SdhD gene. This gene encodes one of the subunits of the mitochondrial complex II (MCII). In this study, we demonstrate that, in contrast to what has been previously established, survival of HSC, and also myeloid and B-lymphoid progenitors, depends on proper mitochondrial activity. In addition, gene expression analysis of these hematopoietic lineages in SDHD-ESR mutants calls into question the proposed activation of Hif1α in response to MCII dysfunction.


Subject(s)
Electron Transport Complex II/metabolism , Gene Deletion , Hematopoietic Stem Cells/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Animals , B-Lymphocytes/immunology , Bone Marrow/metabolism , Cell Hypoxia , Cell Lineage , Cell Survival , Colony-Forming Units Assay , Gene Expression Regulation , Leukocytes/metabolism , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spleen/cytology , Succinate Dehydrogenase , T-Lymphocytes/immunology , Thymus Gland/pathology
11.
J Cell Sci ; 129(24): 4449-4454, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27872153

ABSTRACT

As time passes, mutations accumulate in the genomes of all living organisms. These changes promote genetic diversity, but also precipitate ageing and the initiation of cancer. Food is a common source of mutagens, but little is known about how nutritional factors cause lasting genetic changes in the consuming organism. Here, we describe an unusual genetic interaction between DNA repair in the unicellular amoeba Dictyostelium discoideum and its natural bacterial food source. We found that Dictyostelium deficient in the DNA repair nuclease Xpf (xpf-) display a severe and specific growth defect when feeding on bacteria. Despite being proficient in the phagocytosis and digestion of bacteria, over time, xpf- Dictyostelium feeding on bacteria cease to grow and in many instances die. The Xpf nuclease activity is required for sustained growth using a bacterial food source. Furthermore, the ingestion of this food source leads to a striking accumulation of mutations in the genome of xpf- Dictyostelium This work therefore establishes Dictyostelium as a model genetic system to dissect nutritional genotoxicity, providing insight into how phagocytosis can induce mutagenesis and compromise survival fitness.


Subject(s)
Dictyostelium/metabolism , Mutagenesis , Phagocytosis , Protozoan Proteins/metabolism , Amino Acid Sequence , DNA Repair/genetics , Dictyostelium/cytology , Dictyostelium/growth & development , Phagocytosis/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
12.
PLoS Genet ; 11(11): e1005674, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26584049

ABSTRACT

Co-transcriptional RNA-DNA hybrids (R loops) cause genome instability. To prevent harmful R loop accumulation, cells have evolved specific eukaryotic factors, one being the BRCA2 double-strand break repair protein. As BRCA2 also protects stalled replication forks and is the FANCD1 member of the Fanconi Anemia (FA) pathway, we investigated the FA role in R loop-dependent genome instability. Using human and murine cells defective in FANCD2 or FANCA and primary bone marrow cells from FANCD2 deficient mice, we show that the FA pathway removes R loops, and that many DNA breaks accumulated in FA cells are R loop-dependent. Importantly, FANCD2 foci in untreated and MMC-treated cells are largely R loop dependent, suggesting that the FA functions at R loop-containing sites. We conclude that co-transcriptional R loops and R loop-mediated DNA damage greatly contribute to genome instability and that one major function of the FA pathway is to protect cells from R loops.


Subject(s)
BRCA2 Protein/genetics , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Genomic Instability/genetics , Animals , DNA/chemistry , DNA/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , HeLa Cells , Humans , Mice , RNA/chemistry , RNA/genetics
13.
Mol Cell ; 60(1): 177-88, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26412304

ABSTRACT

Endogenous formaldehyde is produced by numerous biochemical pathways fundamental to life, and it can crosslink both DNA and proteins. However, the consequences of its accumulation are unclear. Here we show that endogenous formaldehyde is removed by the enzyme alcohol dehydrogenase 5 (ADH5/GSNOR), and Adh5(-/-) mice therefore accumulate formaldehyde adducts in DNA. The repair of this damage is mediated by FANCD2, a DNA crosslink repair protein. Adh5(-/-)Fancd2(-/-) mice reveal an essential requirement for these protection mechanisms in hematopoietic stem cells (HSCs), leading to their depletion and precipitating bone marrow failure. More widespread formaldehyde-induced DNA damage also causes karyomegaly and dysfunction of hepatocytes and nephrons. Bone marrow transplantation not only rescued hematopoiesis but, surprisingly, also preserved nephron function. Nevertheless, all of these animals eventually developed fatal malignancies. Formaldehyde is therefore an important source of endogenous DNA damage that is counteracted in mammals by a conserved protection mechanism.


Subject(s)
Alcohol Dehydrogenase/metabolism , Carcinogens/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Formaldehyde/metabolism , Mutagens/metabolism , Alcohol Dehydrogenase/genetics , Animals , Cells, Cultured , DNA Adducts/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Gene Knockout Techniques , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Mice
14.
PLoS Genet ; 10(3): e1004205, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24603549

ABSTRACT

Ribosomal protein L3 is an evolutionarily conserved protein that participates in the assembly of early pre-60S particles. We report that the rpl3[W255C] allele, which affects the affinity and function of translation elongation factors, impairs cytoplasmic maturation of 20S pre-rRNA. This was not seen for other mutations in or depletion of L3 or other 60S ribosomal proteins. Surprisingly, pre-40S particles containing 20S pre-rRNA form translation-competent 80S ribosomes, and translation inhibition partially suppresses 20S pre-rRNA accumulation. The GTP-dependent translation initiation factor Fun12 (yeast eIF5B) shows similar in vivo binding to ribosomal particles from wild-type and rpl3[W255C] cells. However, the GTPase activity of eIF5B failed to stimulate processing of 20S pre-rRNA when assayed with ribosomal particles purified from rpl3[W255C] cells. We conclude that L3 plays an important role in the function of eIF5B in stimulating 3' end processing of 18S rRNA in the context of 80S ribosomes that have not yet engaged in translation. These findings indicate that the correct conformation of the GTPase activation region is assessed in a quality control step during maturation of cytoplasmic pre-ribosomal particles.


Subject(s)
Ribosomal Proteins/genetics , Ribosome Subunits, Large, Eukaryotic/genetics , Ribosome Subunits, Small, Eukaryotic/genetics , Saccharomyces cerevisiae/genetics , Alleles , Cytoplasm/genetics , Cytoplasm/metabolism , Eukaryotic Initiation Factors/genetics , Mutation , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , RNA Precursors/genetics , RNA, Ribosomal, 18S/genetics , Ribosomal Protein L3 , Ribosomal Proteins/metabolism , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism
15.
Nat Struct Mol Biol ; 18(12): 1432-4, 2011 Nov 13.
Article in English | MEDLINE | ID: mdl-22081012

ABSTRACT

Metabolism is predicted to generate formaldehyde, a toxic, simple, reactive aldehyde that can damage DNA. Here we report a synthetic lethal interaction in avian cells between ADH5, encoding the main formaldehyde-detoxifying enzyme, and the Fanconi anemia (FA) DNA-repair pathway. These results define a fundamental role for the combined action of formaldehyde catabolism and DNA cross-link repair in vertebrate cell survival.


Subject(s)
DNA Repair , Fanconi Anemia/metabolism , Formaldehyde/metabolism , Aldehyde Oxidoreductases/genetics , Animals , Cell Line , Chickens/genetics , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/physiology , Fanconi Anemia Complementation Group L Protein/genetics , Fanconi Anemia Complementation Group L Protein/physiology , Gene Knockout Techniques , Metabolic Networks and Pathways
16.
Nature ; 475(7354): 53-8, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21734703

ABSTRACT

Reactive aldehydes are common carcinogens. They are also by-products of several metabolic pathways and, without enzymatic catabolism, may accumulate and cause DNA damage. Ethanol, which is metabolised to acetaldehyde, is both carcinogenic and teratogenic in humans. Here we find that the Fanconi anaemia DNA repair pathway counteracts acetaldehyde-induced genotoxicity in mice. Our results show that the acetaldehyde-catabolising enzyme Aldh2 is essential for the development of Fancd2(-/-) embryos. Nevertheless, acetaldehyde-catabolism-competent mothers (Aldh2(+/-)) can support the development of double-mutant (Aldh2(-/-)Fancd2(-/-)) mice. However, these embryos are unusually sensitive to ethanol exposure in utero, and ethanol consumption by postnatal double-deficient mice rapidly precipitates bone marrow failure. Lastly, Aldh2(-/-)Fancd2(-/-) mice spontaneously develop acute leukaemia. Acetaldehyde-mediated DNA damage may critically contribute to the genesis of fetal alcohol syndrome in fetuses, as well as to abnormal development, haematopoietic failure and cancer predisposition in Fanconi anaemia patients.


Subject(s)
Aldehydes/antagonists & inhibitors , Aldehydes/toxicity , Fanconi Anemia Complementation Group D2 Protein/metabolism , Acetaldehyde/metabolism , Acetaldehyde/toxicity , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase, Mitochondrial , Aldehydes/metabolism , Alleles , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/physiopathology , Cell Line , Cell Survival/drug effects , Chickens , Clone Cells/drug effects , DNA Damage/genetics , DNA Repair/genetics , Embryo Loss/chemically induced , Embryo Loss/etiology , Embryo, Mammalian/abnormalities , Embryo, Mammalian/drug effects , Embryo, Mammalian/embryology , Ethanol/metabolism , Ethanol/toxicity , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Fetal Alcohol Spectrum Disorders/etiology , Gene Deletion , Genes, Essential , Hematopoiesis/drug effects , Male , Mice , Mice, Inbred C57BL , Precursor Cell Lymphoblastic Leukemia-Lymphoma/chemically induced , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Pregnancy , Teratogens/metabolism , Teratogens/toxicity , Weaning
17.
Nat Genet ; 43(2): 147-52, 2011 02.
Article in English | MEDLINE | ID: mdl-21240276

ABSTRACT

The evolutionarily conserved SLX4 protein, a key regulator of nucleases, is critical for DNA damage response. SLX4 nuclease complexes mediate repair during replication and can also resolve Holliday junctions formed during homologous recombination. Here we describe the phenotype of the Btbd12 knockout mouse, the mouse ortholog of SLX4, which recapitulates many key features of the human genetic illness Fanconi anemia. Btbd12-deficient animals are born at sub-Mendelian ratios, have greatly reduced fertility, are developmentally compromised and are prone to blood cytopenias. Btbd12(-/-) cells prematurely senesce, spontaneously accumulate damaged chromosomes and are particularly sensitive to DNA crosslinking agents. Genetic complementation reveals a crucial requirement for Btbd12 (also known as Slx4) to interact with the structure-specific endonuclease Xpf-Ercc1 to promote crosslink repair. The Btbd12 knockout mouse therefore establishes a disease model for Fanconi anemia and genetically links a regulator of nuclease incision complexes to the Fanconi anemia DNA crosslink repair pathway.


Subject(s)
Fanconi Anemia/genetics , Recombinases/genetics , Recombinases/physiology , Animals , Cellular Senescence , Cross-Linking Reagents/pharmacology , DNA Damage , Female , Fibroblasts/metabolism , Genetic Complementation Test , Hematopoietic Stem Cells , Humans , Magnetic Resonance Imaging/methods , Male , Mice , Mice, Knockout
18.
Science ; 329(5988): 219-23, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20538911

ABSTRACT

A conserved DNA repair response is defective in the human genetic illness Fanconi anemia (FA). Mutation of some FA genes impairs homologous recombination and error-prone DNA repair, rendering FA cells sensitive to DNA cross-linking agents. We found a genetic interaction between the FA gene FANCC and the nonhomologous end joining (NHEJ) factor Ku70. Disruption of both FANCC and Ku70 suppresses sensitivity to cross-linking agents, diminishes chromosome breaks, and reverses defective homologous recombination. Ku70 binds directly to free DNA ends, committing them to NHEJ repair. We show that purified FANCD2, a downstream effector of the FA pathway, might antagonize Ku70 activity by modifying such DNA substrates. These results reveal a function for the FA pathway in processing DNA ends, thereby diverting double-strand break repair away from abortive NHEJ and toward homologous recombination.


Subject(s)
Antigens, Nuclear/genetics , DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/genetics , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Recombination, Genetic , Animals , Antigens, Nuclear/metabolism , Cell Line , Chickens , Chromosome Breakage , Cross-Linking Reagents/pharmacology , DNA-Binding Proteins/metabolism , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/chemistry , Fanconi Anemia Complementation Group D2 Protein/genetics , Gene Conversion , Genes, Immunoglobulin , Humans , Immunoglobulin M/genetics , Ku Autoantigen , Point Mutation , Recombinant Proteins/metabolism
19.
Nucleic Acids Res ; 37(13): 4360-70, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19465393

ABSTRACT

FANCM, the most highly conserved component of the Fanconi Anaemia (FA) pathway can resolve recombination intermediates and remodel synthetic replication forks. However, it is not known if these activities are relevant to how this conserved protein activates the FA pathway and promotes DNA crosslink repair. Here we use chicken DT40 cells to systematically dissect the function of the helicase and nuclease domains of FANCM. Our studies reveal that these domains contribute distinct roles in the tolerance of crosslinker, UV light and camptothecin-induced DNA damage. Although the complete helicase domain is critical for crosslink repair, a predicted inactivating mutation of the Walker B box domain has no impact on FA pathway associated functions. However, this mutation does result in elevated sister chromatid exchanges (SCE). Furthermore, our genetic dissection indicates that FANCM functions with the Blm helicase to suppress spontaneous SCE events. Overall our results lead us to reappraise the role of helicase domain associated activities of FANCM with respect to the activation of the FA pathway, crosslink repair and in the resolution of recombination intermediates.


Subject(s)
Avian Proteins/chemistry , DNA Helicases/chemistry , DNA Repair , Fanconi Anemia Complementation Group Proteins/chemistry , Sister Chromatid Exchange , Alleles , Amino Acid Motifs , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Cell Line , Chickens , DNA Helicases/genetics , DNA Helicases/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Genetic Complementation Test , Phenotype , Point Mutation , Protein Structure, Tertiary
20.
Nucleic Acids Res ; 35(12): 4203-13, 2007.
Article in English | MEDLINE | ID: mdl-17569673

ABSTRACT

Ribosome synthesis in eukaryotes requires a multitude of trans-acting factors. These factors act at many steps as the pre-ribosomal particles travel from the nucleolus to the cytoplasm. In contrast to the well-studied trans-acting factors, little is known about the contribution of the ribosomal proteins to ribosome biogenesis. Herein, we have analysed the role of ribosomal protein Rpl3p in 60S ribosomal subunit biogenesis. In vivo depletion of Rpl3p results in a deficit in 60S ribosomal subunits and the appearance of half-mer polysomes. This phenotype is likely due to the instability of early and intermediate pre-ribosomal particles, as evidenced by the low steady-state levels of 27SA(3), 27SB(S) and 7S(L/S) precursors. Furthermore, depletion of Rpl3p impairs the nucleocytoplasmic export of pre-60S ribosomal particles. Interestingly, flow cytometry analysis indicates that Rpl3p-depleted cells arrest in the G1 phase. Altogether, we suggest that upon depletion of Rpl3p, early assembly of 60S ribosomal subunits is aborted and subsequent steps during their maturation and export prevented.


Subject(s)
Ribosomal Proteins/physiology , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/physiology , Active Transport, Cell Nucleus , Cell Cycle , Cell Nucleus/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Ribosomal/metabolism , Ribosomes/chemistry , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...