Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 11 de 11
Filtrer
1.
Nat Genet ; 43(2): 138-41, 2011 Feb.
Article de Anglais | MEDLINE | ID: mdl-21240277

RÉSUMÉ

DNA interstrand crosslink repair requires several classes of proteins, including structure-specific endonucleases and Fanconi anemia proteins. SLX4, which coordinates three separate endonucleases, was recently recognized as an important regulator of DNA repair. Here we report the first human individuals found to have biallelic mutations in SLX4. These individuals, who were previously diagnosed as having Fanconi anemia, add SLX4 as an essential component to the FA-BRCA genome maintenance pathway.


Sujet(s)
Anémie de Fanconi/génétique , Recombinases/génétique , Allèles , Camptothécine/pharmacologie , Enfant , Réactifs réticulants/pharmacologie , Réparation de l'ADN , Relation dose-effet des médicaments , Protéines du choc thermique HSC70 , Protéines du choc thermique/composition chimique , Humains , Immunoprécipitation , Mâle , Mitomycine/pharmacologie , Mutation , Phénotype
2.
Mol Cell ; 37(6): 865-78, 2010 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-20347428

RÉSUMÉ

FANCM remodels branched DNA structures and plays essential roles in the cellular response to DNA replication stress. Here, we show that FANCM forms a conserved DNA-remodeling complex with a histone-fold heterodimer, MHF. We find that MHF stimulates DNA binding and replication fork remodeling by FANCM. In the cell, FANCM and MHF are rapidly recruited to forks stalled by DNA interstrand crosslinks, and both are required for cellular resistance to such lesions. In vertebrates, FANCM-MHF associates with the Fanconi anemia (FA) core complex, promotes FANCD2 monoubiquitination in response to DNA damage, and suppresses sister-chromatid exchanges. Yeast orthologs of these proteins function together to resist MMS-induced DNA damage and promote gene conversion at blocked replication forks. Thus, FANCM-MHF is an essential DNA-remodeling complex that protects replication forks from yeast to human.


Sujet(s)
Helicase/métabolisme , ADN/métabolisme , Instabilité du génome , Histone/métabolisme , Pliage des protéines , Multimérisation de protéines , Séquence d'acides aminés , Animaux , Lignée cellulaire , Poulets , ADN/génétique , Altération de l'ADN , Helicase/composition chimique , Helicase/génétique , Réplication de l'ADN , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Évolution moléculaire , Protéines des groupes de complémentation de l'anémie de Fanconi , Humains , Données de séquences moléculaires , Liaison aux protéines , Saccharomyces cerevisiae/génétique , Saccharomyces cerevisiae/métabolisme , Schizosaccharomyces/génétique , Schizosaccharomyces/métabolisme , Alignement de séquences , Échange de chromatides soeurs
3.
PLoS One ; 4(9): e6936, 2009 Sep 07.
Article de Anglais | MEDLINE | ID: mdl-19738907

RÉSUMÉ

Cohesion between sister chromatids is essential for faithful chromosome segregation. In budding yeast, the acetyltransferase Eco1/Ctf7 establishes cohesion during DNA replication in S phase and in response to DNA double strand breaks in G2/M phase. In humans two Eco1 orthologs exist: ESCO1 and ESCO2. Both proteins are required for proper sister chromatid cohesion, but their exact function is unclear at present. Since ESCO2 has been identified as the gene defective in the rare autosomal recessive cohesinopathy Roberts syndrome (RBS), cells from RBS patients can be used to elucidate the role of ESCO2. We investigated for the first time RBS cells in comparison to isogenic controls that stably express V5- or GFP-tagged ESCO2. We show that the sister chromatid cohesion defect in the transfected cell lines is rescued and suggest that ESCO2 is regulated by proteasomal degradation in a cell cycle-dependent manner. In comparison to the corrected cells RBS cells were hypersensitive to the DNA-damaging agents mitomycin C, camptothecin and etoposide, while no particular sensitivity to UV, ionizing radiation, hydroxyurea or aphidicolin was found. The cohesion defect of RBS cells and their hypersensitivity to DNA-damaging agents were not corrected by a patient-derived ESCO2 acetyltransferase mutant (W539G), indicating that the acetyltransferase activity of ESCO2 is essential for its function. In contrast to a previous study on cells from patients with Cornelia de Lange syndrome, another cohesinopathy, RBS cells failed to exhibit excessive chromosome aberrations after irradiation in G2 phase of the cell cycle. Our results point at an S phase-specific role for ESCO2 in the maintenance of genome stability.


Sujet(s)
Acetyltransferases/métabolisme , Protéines chromosomiques nonhistones/métabolisme , Fibroblastes/métabolisme , Troubles de la croissance/diagnostic , Camptothécine/pharmacologie , Protéines du cycle cellulaire/métabolisme , Aberrations des chromosomes , Ségrégation des chromosomes , Malformations/diagnostic , Malformations/génétique , Altération de l'ADN , Étoposide/pharmacologie , Troubles de la croissance/génétique , Humains , Nourrisson , Mâle , Mitomycine/pharmacologie , Inhibiteurs de la synthèse d'acide nucléique/pharmacologie , Échange de chromatides soeurs , Syndrome ,
4.
Hum Mol Genet ; 18(18): 3484-95, 2009 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-19561169

RÉSUMÉ

The Fanconi anemia (FA) core complex member FANCM remodels synthetic replication forks and recombination intermediates. Thus far, only one FA patient with FANCM mutations has been described, but the relevance of these mutations for the FA phenotype is uncertain. To provide further experimental access to the FA-M complementation group we have generated Fancm-deficient mice by deleting exon 2. FANCM deficiency caused hypogonadism in mice and hypersensitivity to cross-linking agents in mouse embryonic fibroblasts (MEFs), thus phenocopying other FA mouse models. However, Fancm(Delta2/Delta2) mice also showed unique features atypical for FA mice, including underrepresentation of female Fancm(Delta2/Delta2) mice and decreased overall and tumor-free survival. This increased cancer incidence may be correlated to the role of FANCM in the suppression of spontaneous sister chromatid exchanges as observed in MEFs. In addition, FANCM appeared to have a stimulatory rather than essential role in FANCD2 monoubiquitination. The FA-M mouse model presented here suggests that FANCM functions both inside and outside the FA core complex to maintain genome stability and to prevent tumorigenesis.


Sujet(s)
Protéines des groupes de complémentation de l'anémie de Fanconi/déficit , Protéines des groupes de complémentation de l'anémie de Fanconi/métabolisme , Allèles , Animaux , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Transformation cellulaire néoplasique/anatomopathologie , Cellules cultivées , Anémie de Fanconi/génétique , Anémie de Fanconi/métabolisme , Anémie de Fanconi/anatomopathologie , Protéines des groupes de complémentation de l'anémie de Fanconi/génétique , Femelle , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Ovaire/malformations , Ovaire/métabolisme , Phénotype , Échange de chromatides soeurs , Taux de survie , Testicule/malformations , Testicule/métabolisme
5.
Blood ; 114(1): 174-80, 2009 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-19423727

RÉSUMÉ

FANCM is a component of the Fanconi anemia (FA) core complex and one FA patient (EUFA867) with biallelic mutations in FANCM has been described. Strikingly, we found that EUFA867 also carries biallelic mutations in FANCA. After correcting the FANCA defect in EUFA867 lymphoblasts, a "clean" FA-M cell line was generated. These cells were hypersensitive to mitomycin C, but unlike cells defective in other core complex members, FANCM(-/-) cells were proficient in monoubiquitinating FANCD2 and were sensitive to the topoisomerase inhibitor camptothecin, a feature shared only with the FA subtype D1 and N. In addition, FANCM(-/-) cells were sensitive to UV light. FANCM and a C-terminal deletion mutant rescued the cross-linker sensitivity of FANCM(-/-) cells, whereas a FANCM ATPase mutant did not. Because both mutants restored the formation of FANCD2 foci, we conclude that FANCM functions in an FA core complex-dependent and -independent manner.


Sujet(s)
Helicase/génétique , Helicase/métabolisme , Protéine du groupe de complémentation D2 de l'anémie de Fanconi/génétique , Protéine du groupe de complémentation D2 de l'anémie de Fanconi/métabolisme , Anémie de Fanconi/génétique , Anémie de Fanconi/métabolisme , Adenosine triphosphatases/génétique , Adenosine triphosphatases/métabolisme , Camptothécine/pharmacologie , Lignée cellulaire tumorale , Réactifs réticulants/pharmacologie , Helicase/déficit , Résistance aux substances/génétique , Résistance aux substances/physiologie , Protéine du groupe de complémentation A de l'anémie de Fanconi/génétique , Protéine du groupe de complémentation A de l'anémie de Fanconi/métabolisme , Expression des gènes , Humains , Mutation , Radiotolérance/génétique , Radiotolérance/physiologie , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Transfection , Ubiquitination/génétique , Rayons ultraviolets
6.
Cell Oncol ; 29(3): 211-8, 2007.
Article de Anglais | MEDLINE | ID: mdl-17452773

RÉSUMÉ

To identify the gene underlying Fanconi anemia (FA) complementation group I we studied informative FA-I families by a genome-wide linkage analysis, which resulted in 4 candidate regions together encompassing 351 genes. Candidates were selected via bioinformatics and data mining on the basis of their resemblance to other FA genes/proteins acting in the FA pathway, such as: degree of evolutionary conservation, presence of nuclear localization signals and pattern of tissue-dependent expression. We found a candidate, KIAA1794 on chromosome 15q25-26, to be mutated in 8 affected individuals previously assigned to complementation group I. Western blots of endogenous FANCI indicated that functionally active KIAA1794 protein is lacking in FA-I individuals. Knock-down of KIAA1794 expression by siRNA in HeLa cells caused excessive chromosomal breakage induced by mitomycin C, a hallmark of FA cells. Furthermore, phenotypic reversion of a patient-derived cell line was associated with a secondary genetic alteration at the KIAA1794 locus. These data add up to two conclusions. First, KIAA1794 is a FA gene. Second, this gene is identical to FANCI, since the patient cell lines found mutated in this study included the reference cell line for group I, EUFA592.


Sujet(s)
Protéines des groupes de complémentation de l'anémie de Fanconi/génétique , Adolescent , Adulte , Séquence nucléotidique , Lignée cellulaire , Enfant , Instabilité des chromosomes/génétique , Protéine du groupe de complémentation D2 de l'anémie de Fanconi/métabolisme , Femelle , Génome humain/génétique , Cellules HeLa , Humains , Mâle , Données de séquences moléculaires , Mutation/génétique , Pedigree , Phénotype , Ubiquitine/métabolisme
7.
Blood ; 108(6): 2072-80, 2006 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-16720839

RÉSUMÉ

Fanconi anemia (FA) is a genomic instability disorder, clinically characterized by congenital abnormalities, progressive bone marrow failure, and predisposition to malignancy. Cells derived from patients with FA display a marked sensitivity to DNA cross-linking agents, such as mitomycin C (MMC). This observation has led to the hypothesis that the proteins defective in FA are involved in the sensing or repair of interstrand cross-link lesions of the DNA. A nuclear complex consisting of a majority of the FA proteins plays a crucial role in this process and is required for the monoubiquitination of a downstream target, FANCD2. Two new FA genes, FANCB and FANCL, have recently been identified, and their discovery has allowed a more detailed study into the molecular architecture of the FA pathway. We demonstrate a direct interaction between FANCB and FANCL and that a complex of these proteins binds FANCA. The interaction between FANCA and FANCL is dependent on FANCB, FANCG, and FANCM, but independent of FANCC, FANCE, and FANCF. These findings provide a framework for the protein interactions that occur "upstream" in the FA pathway and suggest that besides the FA core complex different subcomplexes exist that may have specific functions other than the monoubiquitination of FANCD2.


Sujet(s)
Protéines des groupes de complémentation de l'anémie de Fanconi/métabolisme , Anémie de Fanconi/métabolisme , Lignée cellulaire , Anémie de Fanconi/étiologie , Anémie de Fanconi/génétique , Protéine du groupe de complémentation A de l'anémie de Fanconi/composition chimique , Protéine du groupe de complémentation A de l'anémie de Fanconi/génétique , Protéine du groupe de complémentation A de l'anémie de Fanconi/métabolisme , Protéine du groupe de complémentation G de l'anémie de Fanconi/composition chimique , Protéine du groupe de complémentation G de l'anémie de Fanconi/génétique , Protéine du groupe de complémentation G de l'anémie de Fanconi/métabolisme , Protéine du groupe de complémentation L de l'anémie de Fanconi/composition chimique , Protéine du groupe de complémentation L de l'anémie de Fanconi/génétique , Protéine du groupe de complémentation L de l'anémie de Fanconi/métabolisme , Protéines des groupes de complémentation de l'anémie de Fanconi/composition chimique , Protéines des groupes de complémentation de l'anémie de Fanconi/génétique , Humains , Techniques in vitro , Modèles moléculaires , Complexes multiprotéiques , Mutation faux-sens , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Transfection
8.
DNA Repair (Amst) ; 5(5): 556-65, 2006 May 10.
Article de Anglais | MEDLINE | ID: mdl-16513431

RÉSUMÉ

The Fanconi anemia (FA) protein FANCE is an essential component of the nuclear FA core complex, which is required for monoubiquitination of the downstream target FANCD2, an important step in the FA pathway of DNA cross-link repair. FANCE is predominantly localized in the nucleus and acts as a molecular bridge between the FA core complex and FANCD2, through direct binding of both FANCC and FANCD2. At present, it is poorly understood how the nuclear accumulation of FANCE is regulated and therefore we investigated the nuclear localization of this FA protein. We found that FANCE has a strong tendency to localize in the nucleus, since the addition of a nuclear export signal does not interfere with the nuclear localization of FANCE. We also demonstrate that the nuclear accumulation of FANCE does not rely solely on its nuclear localization signal motifs, but also on FANCC. The other FA proteins are not involved in the nuclear accumulation of FANCE, indicating a tight relationship between FANCC and FANCE, as suggested from their direct interaction. Finally, we show that the region of FANCE interacting with FANCC appears to be different from the region involved in binding FANCD2. This strengthens the idea that FANCE recruits FANCD2 to the core complex, without interfering with the binding of FANCC.


Sujet(s)
Protéine du groupe de complémentation C de l'anémie de Fanconi/métabolisme , Protéine du groupe de complémentation E de l'anémie de Fanconi/métabolisme , Transport nucléaire actif , Séquence d'acides aminés , Sites de fixation , Lignée cellulaire , Anémie de Fanconi/génétique , Anémie de Fanconi/métabolisme , Protéine du groupe de complémentation C de l'anémie de Fanconi/composition chimique , Protéine du groupe de complémentation C de l'anémie de Fanconi/génétique , Protéine du groupe de complémentation E de l'anémie de Fanconi/composition chimique , Protéine du groupe de complémentation E de l'anémie de Fanconi/génétique , Cellules HeLa , Humains , Mutagenèse dirigée , Signaux d'export nucléaire/génétique , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Transfection , Techniques de double hybride
9.
Nat Genet ; 37(9): 958-63, 2005 Sep.
Article de Anglais | MEDLINE | ID: mdl-16116422

RÉSUMÉ

Fanconi anemia is a genetic disease characterized by genomic instability and cancer predisposition. Nine genes involved in Fanconi anemia have been identified; their products participate in a DNA damage-response network involving BRCA1 and BRCA2 (refs. 2,3). We previously purified a Fanconi anemia core complex containing the FANCL ubiquitin ligase and six other Fanconi anemia-associated proteins. Each protein in this complex is essential for monoubiquitination of FANCD2, a key reaction in the Fanconi anemia DNA damage-response pathway. Here we show that another component of this complex, FAAP250, is mutant in individuals with Fanconi anemia of a new complementation group (FA-M). FAAP250 or FANCM has sequence similarity to known DNA-repair proteins, including archaeal Hef, yeast MPH1 and human ERCC4 or XPF. FANCM can dissociate DNA triplex, possibly owing to its ability to translocate on duplex DNA. FANCM is essential for monoubiquitination of FANCD2 and becomes hyperphosphorylated in response to DNA damage. Our data suggest an evolutionary link between Fanconi anemia-associated proteins and DNA repair; FANCM may act as an engine that translocates the Fanconi anemia core complex along DNA.


Sujet(s)
Archéobactéries/composition chimique , Helicase/génétique , Réparation de l'ADN , Anémie de Fanconi/génétique , Hémagglutinines virales/génétique , Ligases/génétique , Protéines de fusion virale/génétique , Protéine BRCA1/génétique , Protéine BRCA2/génétique , Évolution biologique , ADN/métabolisme , Helicase/déficit , Helicase/métabolisme , Anémie de Fanconi/enzymologie , Protéine du groupe de complémentation D2 de l'anémie de Fanconi , Protéine du groupe de complémentation L de l'anémie de Fanconi , Humains , Immunoprécipitation , Ligases/déficit , Ligases/métabolisme , Données de séquences moléculaires , Mutation , Protéines nucléaires/métabolisme , Phosphorylation , Transport des protéines , Ubiquitine/métabolisme , Protéines de fusion virale/déficit
10.
Nat Genet ; 37(9): 934-5, 2005 Sep.
Article de Anglais | MEDLINE | ID: mdl-16116423

RÉSUMÉ

The protein predicted to be defective in individuals with Fanconi anemia complementation group J (FA-J), FANCJ, is a missing component in the Fanconi anemia pathway of genome maintenance. Here we identify pathogenic mutations in eight individuals with FA-J in the gene encoding the DEAH-box DNA helicase BRIP1, also called FANCJ. This finding is compelling evidence that the Fanconi anemia pathway functions through a direct physical interaction with DNA.


Sujet(s)
Chromosomes humains de la paire 17 , Protéines de liaison à l'ADN/déficit , Protéines de liaison à l'ADN/génétique , Anémie de Fanconi/génétique , Mutation/génétique , RNA helicases/déficit , RNA helicases/génétique , Protéines des groupes de complémentation de l'anémie de Fanconi , Test de complémentation , Humains , Répétitions microsatellites , Données de séquences moléculaires , Délétion de séquence
11.
Blood ; 103(7): 2498-503, 2004 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-14630800

RÉSUMÉ

Fanconi anemia (FA) is an autosomal recessive syndrome featuring diverse symptoms including progressive bone marrow failure and early occurrence of acute myeloid leukemia. Nine genetic subtypes have been described for FA (A, B, C, D1, D2, E, F, G, and L), all of which have been connected to distinct disease genes, except B. Here we report on 8 unrelated FA patients who were excluded from the known subtypes on the basis of phenotypic correction or genetic data. Four of these cell lines failed to complement each other in somatic cell hybrids and therefore represent a new group, termed FA-I. The remaining cell lines complemented group FA-I but did not complement each other, thus representing a second new group, FA-J. Both FA-I and -J cell lines were capable of forming an FA multiprotein core complex. This complex is required for activation of the FANCD2 protein by mono-ubiquitination, a key downstream event in the FA pathway. In FA-I cells FANCD2 was not mono-ubiquitinated, indicating a defect upstream in the FA pathway, whereas in FA-J cells FANCD2 was mono-ubiquitinated, indicating a downstream defect. Our results suggest that the FA pathway of genome stabilization may be controlled by at least 11 different genes, including FANCI and FANCJ.


Sujet(s)
Anémie de Fanconi/classification , Anémie de Fanconi/génétique , Polymorphisme génétique , Division cellulaire , Fusion cellulaire , Lignée cellulaire , Enfant , Enfant d'âge préscolaire , Anémie de Fanconi/anatomopathologie , Gènes récessifs , Test de complémentation , Humains , Transfection
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE