Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nucleic Acids Res ; 48(5): 2442-2456, 2020 03 18.
Article in English | MEDLINE | ID: mdl-31960047

ABSTRACT

The tumor suppressor BRCA2 is essential for homologous recombination (HR), replication fork stability and DNA interstrand crosslink (ICL) repair in vertebrates. We show that ectopic production of HSF2BP, a BRCA2-interacting protein required for meiotic HR during mouse spermatogenesis, in non-germline human cells acutely sensitize them to ICL-inducing agents (mitomycin C and cisplatin) and PARP inhibitors, resulting in a phenotype characteristic of cells from Fanconi anemia (FA) patients. We biochemically recapitulate the suppression of ICL repair and establish that excess HSF2BP compromises HR by triggering the removal of BRCA2 from the ICL site and thereby preventing the loading of RAD51. This establishes ectopic expression of a wild-type meiotic protein in the absence of any other protein-coding mutations as a new mechanism that can lead to an FA-like cellular phenotype. Naturally occurring elevated production of HSF2BP in tumors may be a source of cancer-promoting genomic instability and also a targetable vulnerability.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , DNA Repair , Heat-Shock Proteins/metabolism , Homologous Recombination , Animals , BRCA2 Protein/metabolism , Cell Line , DNA Damage , Fanconi Anemia/genetics , Humans , Mice , Protein Binding , Proteolysis , Rad51 Recombinase/metabolism , Xenopus
2.
Mol Cell ; 47(1): 61-75, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22705371

ABSTRACT

The Fanconi anemia (FA) protein network is necessary for repair of DNA interstrand crosslinks (ICLs), but its control mechanism remains unclear. Here we show that the network is regulated by a ubiquitin signaling cascade initiated by RNF8 and its partner, UBC13, and mediated by FAAP20, a component of the FA core complex. FAAP20 preferentially binds the ubiquitin product of RNF8-UBC13, and this ubiquitin-binding activity and RNF8-UBC13 are both required for recruitment of FAAP20 to ICLs. Both RNF8 and FAAP20 are required for recruitment of FA core complex and FANCD2 to ICLs, whereas RNF168 can modulate efficiency of the recruitment. RNF8 and FAAP20 are needed for efficient FANCD2 monoubiquitination, a key step of the FA network; RNF8 and the FA core complex work in the same pathway to promote cellular resistance to ICLs. Thus, the RNF8-FAAP20 ubiquitin cascade is critical for recruiting FA core complex to ICLs and for normal function of the FA network.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Ubiquitination , Amino Acid Sequence , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Lysine/chemistry , Lysine/genetics , Lysine/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Protein Binding , Protein Structure, Tertiary , RNA Interference , Sequence Homology, Amino Acid , Signal Transduction , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
3.
Mol Cell ; 37(6): 865-78, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20347428

ABSTRACT

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.


Subject(s)
DNA Helicases/metabolism , DNA/metabolism , Genomic Instability , Histones/metabolism , Protein Folding , Protein Multimerization , Amino Acid Sequence , Animals , Cell Line , Chickens , DNA/genetics , DNA Damage , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Evolution, Molecular , Fanconi Anemia Complementation Group Proteins , Humans , Molecular Sequence Data , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Sequence Alignment , Sister Chromatid Exchange
4.
J Oral Pathol Med ; 45(3): 189-92, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26306924

ABSTRACT

OBJECTIVE: Oral squamous cell carcinoma (OSCC) may develop in young adults. In contrast to older patients, the well-known etiological factors, exposure to tobacco and alcohol, play a minor role in the carcinogenesis in this patient group. It has been suggested that an intrinsic susceptibility to environmental genotoxic exposures plays a role in the development of OSCC in these patients. The hypothesis was tested whether young OSCC patients have an increased sensitivity to induced chromosomal damage. SUBJECTS AND METHODS: Fourteen OSCC patients with an average age of 32 years (range 20-42) were selected. Peripheral blood lymphocytes and skin fibroblasts of patients and 14 healthy controls were subjected to the chromosome breakage test with Mitomycin C. This test is routinely used to identify Fanconi anemia patients, who are well-known for their inherited high sensitivity to this type of DNA damage, but also for the high risk to develop OSCC. Human papilloma virus status of the carcinomas was also determined. RESULTS: None of the 14 young patients with OSCC had an increased response in the MMC-chromosomal breakage test. All tumors tested negative for human papilloma virus. CONCLUSION: No evidence was obtained for the existence of a constitutional hypersensitivity to DNA chromosomal damage as a potential risk factor for OSCC in young adults.


Subject(s)
Carcinoma, Squamous Cell/genetics , Chromosome Breakage , Head and Neck Neoplasms/genetics , Mouth Neoplasms/genetics , Adult , Carcinoma, Squamous Cell/blood , DNA Damage , Fanconi Anemia/genetics , Female , Genetic Predisposition to Disease , Head and Neck Neoplasms/blood , Humans , Male , Mitomycin/pharmacology , Mouth Neoplasms/blood , Papillomaviridae , Risk Factors , Squamous Cell Carcinoma of Head and Neck , Young Adult
5.
Am J Hum Genet ; 86(2): 262-6, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20137776

ABSTRACT

The iron-sulfur-containing DNA helicases XPD, FANCJ, DDX11, and RTEL represent a small subclass of superfamily 2 helicases. XPD and FANCJ have been connected to the genetic instability syndromes xeroderma pigmentosum and Fanconi anemia. Here, we report a human individual with biallelic mutations in DDX11. Defective DDX11 is associated with a unique cellular phenotype in which features of Fanconi anemia (drug-induced chromosomal breakage) and Roberts syndrome (sister chromatid cohesion defects) coexist. The DDX11-deficient patient represents another cohesinopathy, besides Cornelia de Lange syndrome and Roberts syndrome, and shows that DDX11 functions at the interface between DNA repair and sister chromatid cohesion.


Subject(s)
Abnormalities, Multiple/enzymology , Abnormalities, Multiple/genetics , Chromosome Breakage , DEAD-box RNA Helicases/genetics , DNA Helicases/genetics , Mutation/genetics , Sister Chromatid Exchange/genetics , Xeroderma Pigmentosum/genetics , Adolescent , Base Sequence , Child, Preschool , DEAD-box RNA Helicases/deficiency , DNA Helicases/deficiency , DNA Mutational Analysis , Female , Humans , Infant , Infant, Newborn , Male , Molecular Sequence Data , Neoplasms/genetics , Pedigree , Phenotype , Poland , Pregnancy , Syndrome
6.
J Pathol ; 226(1): 28-39, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21915857

ABSTRACT

Fanconi anaemia (FA) is a rare recessive disorder marked by developmental abnormalities, bone marrow failure, and a high risk for the development of leukaemia and solid tumours. The inactivation of FA genes, in particular FANCF, has also been documented in sporadic tumours in non-FA patients. To study whether there is a causal relationship between FA pathway defects and tumour development, we have generated a mouse model with a targeted disruption of the FA core complex gene Fancf. Fancf-deficient mouse embryonic fibroblasts displayed a phenotype typical for FA cells: they showed an aberrant response to DNA cross-linking agents as manifested by G(2) arrest, chromosomal aberrations, reduced survival, and an inability to monoubiquitinate FANCD2. Fancf homozygous mice were viable, born following a normal Mendelian distribution, and showed no growth retardation or developmental abnormalities. The gonads of Fancf mutant mice functioned abnormally, showing compromised follicle development and spermatogenesis as has been observed in other FA mouse models and in FA patients. In a cohort of Fancf-deficient mice, we observed decreased overall survival and increased tumour incidence. Notably, in seven female mice, six ovarian tumours developed: five granulosa cell tumours and one luteoma. One mouse had developed tumours in both ovaries. High-resolution array comparative genomic hybridization (aCGH) on these tumours suggests that the increased incidence of ovarian tumours correlates with the infertility in Fancf-deficient mice and the genomic instability characteristic of FA pathway deficiency.


Subject(s)
Fanconi Anemia Complementation Group F Protein/genetics , Granulosa Cell Tumor/genetics , Luteoma/genetics , Ovarian Neoplasms/genetics , Animals , Comparative Genomic Hybridization , Disease Models, Animal , Fanconi Anemia Complementation Group F Protein/deficiency , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Nat Genet ; 35(2): 165-70, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12973351

ABSTRACT

Fanconi anemia is a recessively inherited disease characterized by congenital defects, bone marrow failure and cancer susceptibility. Cells from individuals with Fanconi anemia are highly sensitive to DNA-crosslinking drugs, such as mitomycin C (MMC). Fanconi anemia proteins function in a DNA damage response pathway involving breast cancer susceptibility gene products, BRCA1 and BRCA2 (refs. 1,2). A key step in this pathway is monoubiquitination of FANCD2, resulting in the redistribution of FANCD2 to nuclear foci containing BRCA1 (ref. 3). The underlying mechanism is unclear because the five Fanconi anemia proteins known to be required for this ubiquitination have no recognizable ubiquitin ligase motifs. Here we report a new component of a Fanconi anemia protein complex, called PHF9, which possesses E3 ubiquitin ligase activity in vitro and is essential for FANCD2 monoubiquitination in vivo. Because PHF9 is defective in a cell line derived from an individual with Fanconi anemia, we conclude that PHF9 (also called FANCL) represents a novel Fanconi anemia complementation group (FA-L). Our data suggest that PHF9 has a crucial role in the Fanconi anemia pathway as the likely catalytic subunit required for monoubiquitination of FANCD2.


Subject(s)
Fanconi Anemia/genetics , Ligases/genetics , Nuclear Proteins/genetics , Sequence Deletion , Amino Acid Sequence , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Base Sequence , Chromosome Aberrations , Fanconi Anemia/enzymology , Fanconi Anemia Complementation Group D2 Protein , Fanconi Anemia Complementation Group L Protein , Humans , Ligases/deficiency , Molecular Sequence Data , Sequence Alignment , Sequence Homology, Amino Acid , Ubiquitin/metabolism
8.
Sci Adv ; 8(3): eabk0114, 2022 01 21.
Article in English | MEDLINE | ID: mdl-35044816

ABSTRACT

Budding uninhibited by benzimidazoles (BUB1) contributes to multiple mitotic processes. Here, we describe the first two patients with biallelic BUB1 germline mutations, who both display microcephaly, intellectual disability, and several patient-specific features. The identified mutations cause variable degrees of reduced total protein level and kinase activity, leading to distinct mitotic defects. Both patients' cells show prolonged mitosis duration, chromosome segregation errors, and an overall functional spindle assembly checkpoint. However, while BUB1 levels mostly affect BUBR1 kinetochore recruitment, impaired kinase activity prohibits centromeric recruitment of Aurora B, SGO1, and TOP2A, correlating with anaphase bridges, aneuploidy, and defective sister chromatid cohesion. We do not observe accelerated cohesion fatigue. We hypothesize that unresolved DNA catenanes increase cohesion strength, with concomitant increase in anaphase bridges. In conclusion, BUB1 mutations cause a neurodevelopmental disorder, with clinical and cellular phenotypes that partially resemble previously described syndromes, including autosomal recessive primary microcephaly, mosaic variegated aneuploidy, and cohesinopathies.


Subject(s)
Chromosome Segregation , Microcephaly , Aneuploidy , Chromosome Segregation/genetics , Humans , Microcephaly/genetics , Mutation , Protein Serine-Threonine Kinases/genetics
9.
Hum Mol Genet ; 18(18): 3484-95, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19561169

ABSTRACT

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.


Subject(s)
Fanconi Anemia Complementation Group Proteins/deficiency , Fanconi Anemia Complementation Group Proteins/metabolism , Alleles , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group Proteins/genetics , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovary/abnormalities , Ovary/metabolism , Phenotype , Sister Chromatid Exchange , Survival Rate , Testis/abnormalities , Testis/metabolism
10.
PLoS One ; 15(1): e0220348, 2020.
Article in English | MEDLINE | ID: mdl-31935221

ABSTRACT

In a process linked to DNA replication, duplicated chromosomes are entrapped in large, circular cohesin complexes and functional sister chromatid cohesion (SCC) is established by acetylation of the SMC3 cohesin subunit. Roberts Syndrome (RBS) and Warsaw Breakage Syndrome (WABS) are rare human developmental syndromes that are characterized by defective SCC. RBS is caused by mutations in the SMC3 acetyltransferase ESCO2, whereas mutations in the DNA helicase DDX11 lead to WABS. We found that WABS-derived cells predominantly rely on ESCO2, not ESCO1, for residual SCC, growth and survival. Reciprocally, RBS-derived cells depend on DDX11 to maintain low levels of SCC. Synthetic lethality between DDX11 and ESCO2 correlated with a prolonged delay in mitosis, and was rescued by knockdown of the cohesin remover WAPL. Rescue experiments using human or mouse cDNAs revealed that DDX11, ESCO1 and ESCO2 act on different but related aspects of SCC establishment. Furthermore, a DNA binding DDX11 mutant failed to correct SCC in WABS cells and DDX11 deficiency reduced replication fork speed. We propose that DDX11, ESCO1 and ESCO2 control different fractions of cohesin that are spatially and mechanistically separated.


Subject(s)
Acetyltransferases/genetics , Cell Cycle Proteins/genetics , Chromatids/metabolism , Chromosomal Proteins, Non-Histone/genetics , DEAD-box RNA Helicases/genetics , DNA Helicases/genetics , Epithelial Cells/enzymology , Fibroblasts/enzymology , Acetyltransferases/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Transformed , Cell Proliferation , Chromatids/ultrastructure , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Breakage , Chromosome Segregation , Craniofacial Abnormalities/enzymology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , DEAD-box RNA Helicases/metabolism , DNA Helicases/metabolism , Ectromelia/enzymology , Ectromelia/genetics , Ectromelia/pathology , Epithelial Cells/pathology , Fibroblasts/pathology , Gene Expression , Humans , Hypertelorism/enzymology , Hypertelorism/genetics , Hypertelorism/pathology , Mice , Mitosis , Models, Biological , Mutation , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Cohesins
11.
Dev Cell ; 52(6): 683-698.e7, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32084359

ABSTRACT

Premature loss of sister chromatid cohesion at metaphase is a diagnostic marker for different cohesinopathies. Here, we report that metaphase spreads of many cancer cell lines also show premature loss of sister chromatid cohesion. Cohesion loss occurs independently of mutations in cohesion factors including SA2, a cohesin subunit frequently inactivated in cancer. In untransformed cells, induction of DNA replication stress by activation of oncogenes or inhibition of DNA replication is sufficient to trigger sister chromatid cohesion loss. Importantly, cell growth under conditions of replication stress requires the cohesin remover WAPL. WAPL promotes rapid RAD51-dependent repair and restart of broken replication forks. We propose that active removal of cohesin allows cancer cells to overcome DNA replication stress. This leads to oncogene-induced cohesion loss from newly synthesized sister chromatids that may contribute to genomic instability and likely represents a targetable cancer cell vulnerability.


Subject(s)
Carrier Proteins/metabolism , Chromatids/genetics , DNA Repair , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , ras Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , Chromosomal Proteins, Non-Histone/metabolism , DNA Replication , HEK293 Cells , Humans , Mice , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Cohesins
12.
Nat Commun ; 11(1): 4287, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32855419

ABSTRACT

Warsaw Breakage Syndrome (WABS) is a rare disorder related to cohesinopathies and Fanconi anemia, caused by bi-allelic mutations in DDX11. Here, we report multiple compound heterozygous WABS cases, each displaying destabilized DDX11 protein and residual DDX11 function at the cellular level. Patient-derived cell lines exhibit sensitivity to topoisomerase and PARP inhibitors, defective sister chromatid cohesion and reduced DNA replication fork speed. Deleting DDX11 in RPE1-TERT cells inhibits proliferation and survival in a TP53-dependent manner and causes chromosome breaks and cohesion defects, independent of the expressed pseudogene DDX12p. Importantly, G-quadruplex (G4) stabilizing compounds induce chromosome breaks and cohesion defects which are strongly aggravated by inactivation of DDX11 but not FANCJ. The DNA helicase domain of DDX11 is essential for sister chromatid cohesion and resistance to G4 stabilizers. We propose that DDX11 is a DNA helicase protecting against G4 induced double-stranded breaks and concomitant loss of cohesion, possibly at DNA replication forks.


Subject(s)
Abnormalities, Multiple/etiology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , G-Quadruplexes , Sister Chromatid Exchange , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Cell Proliferation , DEAD-box RNA Helicases/chemistry , DNA Helicases/chemistry , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Humans , Male , Middle Aged , Mutation, Missense , Protein Stability , Pseudogenes , RNA Helicases/genetics , RNA Helicases/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Syndrome , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Cell Oncol ; 29(3): 211-8, 2007.
Article in English | MEDLINE | ID: mdl-17452773

ABSTRACT

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.


Subject(s)
Fanconi Anemia Complementation Group Proteins/genetics , Adolescent , Adult , Base Sequence , Cell Line , Child , Chromosomal Instability/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Female , Genome, Human/genetics , HeLa Cells , Humans , Male , Molecular Sequence Data , Mutation/genetics , Pedigree , Phenotype , Ubiquitin/metabolism
14.
Cancer Res ; 63(10): 2688-94, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12750298

ABSTRACT

A 2-year old boy was diagnosed with Fanconi anemia (FA) and acute myeloid leukemia (AML). A cell line (termed FA-AML1) was established from blast cells obtained after a second relapse after a successful bone marrow transplant. Histochemical and surface marker analysis confirmed that the cells were derived from the myeloid lineage. Cytogenetic analysis revealed multiple chromosomal aberrations, including a ring 7. Stable proliferation of the cultured cells was absolutely dependent on the presence of granulocyte macrophage colony-stimulating factor or interleukin 3. This is the first AML cell line successfully established from a FA patient. Remarkably, FA-AML1 cells appeared to lack the characteristic cellular FA phenotype, i.e., a hypersensitivity to growth inhibition and chromosomal breakage by the cross-linking agent mitomycin C. Genomic DNA from the patient showed biallelic mutations [8415G>T (K2729N)and 8732C>A (S2835STOP)] in the breast cancer susceptibility gene FANCD1/BRCA2 [N. Howlett et al., Science (Wash. DC), 297: 606-609, 2002]. In the AML cells, however, the 8732C>A nonsense mutation was changed into a missense mutation by a secondary alteration, 8731T>G, resulting in 2835E, which restored the open-reading frame of the gene and could explain the reverted phenotype of these cells. Loss of the FA phenotype by genetic correction of a FA gene mutation during AML progression may be a common late event in the pathogenesis of AML in FA patients, which may be treatment related. This finding suggests a novel mechanistic principle of tumor progression based on the genetic correction of an early caretaker gene defect.


Subject(s)
Fanconi Anemia/genetics , Genes, BRCA2 , Leukemia, Myeloid, Acute/genetics , Mutation , Tumor Cells, Cultured , Alleles , Antigens, CD/biosynthesis , Cell Division/drug effects , Child, Preschool , Fanconi Anemia/complications , Fanconi Anemia/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-3/pharmacology , Karyotyping , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/pathology , Male
15.
Free Radic Biol Med ; 89: 863-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26456054

ABSTRACT

Aim of this study was to explore cellular changes associated with increased resistance to atmospheric oxygen using high-resolution DNA and RNA profiling combined with functional studies. Two independently selected oxygen-resistant substrains of HeLa cells (capable of proliferating at >80% O2, i.e. hyperoxia) were compared with their parental cells (adapted to growth at 20% O2, but unable to grow at >80% O2). A striking consistent alteration found to be associated with the oxygen-resistant state appeared to be an amplified and overexpressed region on chromosome 16p13.3 harboring 21 genes. The driver gene of this amplification was identified by functional studies as NARFL, which encodes a component of the cytosolic iron-sulfur cluster assembly system. In line with this result we found the cytosolic c-aconitase activity as well as the nuclear protein RTEL1, both Fe-S dependent proteins, to be protected by NARFL overexpression under hyperoxia. In addition, we observed a protective effect of NARFL against hyperoxia-induced loss of sister-chromatid cohesion. NARFL thus appeared to be a key factor in the cellular defense against hyperoxia-induced oxidative stress in human cells. Our findings suggest that new insight into age-related degenerative processes may come from studies that specifically address the involvement of iron-sulfur proteins.


Subject(s)
Chromosome Breakage , Hydrogenase/metabolism , Iron-Sulfur Proteins/metabolism , Oxidative Stress/physiology , Aconitate Hydratase/metabolism , Blotting, Western , Cell Survival , Cytosol/metabolism , DNA Helicases/metabolism , HeLa Cells , Humans , Hydrogenase/antagonists & inhibitors , Hydrogenase/genetics , Iron-Sulfur Proteins/genetics , Mitochondria/metabolism , Protein Interaction Domains and Motifs , RNA, Small Interfering/genetics , Transcriptome
16.
Cancer Res ; 75(17): 3543-53, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26122845

ABSTRACT

Failure to repair DNA damage or defective sister chromatid cohesion, a process essential for correct chromosome segregation, can be causative of chromosomal instability (CIN), which is a hallmark of many types of cancers. We investigated how frequent this occurs in head and neck squamous cell carcinoma (HNSCC) and whether specific mechanisms or genes could be linked to these phenotypes. The genomic instability syndrome Fanconi anemia is caused by mutations in any of at least 16 genes regulating DNA interstrand crosslink (ICL) repair. Since patients with Fanconi anemia have a high risk to develop HNSCC, we investigated whether and to which extent Fanconi anemia pathway inactivation underlies CIN in HNSCC of non-Fanconi anemia individuals. We observed ICL-induced chromosomal breakage in 9 of 17 (53%) HNSCC cell lines derived from patients without Fanconi anemia. In addition, defective sister chromatid cohesion was observed in five HNSCC cell lines. Inactivation of FANCM was responsible for chromosomal breakage in one cell line, whereas in two other cell lines, somatic mutations in PDS5A or STAG2 resulted in inadequate sister chromatid cohesion. In addition, FANCF methylation was found in one cell line by screening an additional panel of 39 HNSCC cell lines. Our data demonstrate that CIN in terms of ICL-induced chromosomal breakage and defective chromatid cohesion is frequently observed in HNSCC. Inactivation of known Fanconi anemia and chromatid cohesion genes does explain CIN in the minority of cases. These findings point to phenotypes that may be highly relevant in treatment response of HNSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Chromosomal Instability/genetics , Fanconi Anemia/genetics , Head and Neck Neoplasms/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Chromatids/genetics , DNA Damage/genetics , DNA Repair/genetics , Fanconi Anemia/pathology , Female , Head and Neck Neoplasms/pathology , Humans , Male , Mutation , Neoplasm Staging , Sister Chromatid Exchange , Squamous Cell Carcinoma of Head and Neck
17.
Nat Commun ; 6: 8399, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26423134

ABSTRACT

Warsaw breakage syndrome (WABS) is caused by defective DDX11, a DNA helicase that is essential for chromatid cohesion. Here, a paired genome-wide siRNA screen in patient-derived cell lines reveals that WABS cells do not tolerate partial depletion of individual APC/C subunits or the spindle checkpoint inhibitor p31(comet). A combination of reduced cohesion and impaired APC/C function also leads to fatal mitotic arrest in diploid RPE1 cells. Moreover, WABS cell lines, and several cancer cell lines with cohesion defects, display a highly increased response to a new cell-permeable APC/C inhibitor, apcin, but not to the spindle poison paclitaxel. Synthetic lethality of APC/C inhibition and cohesion defects strictly depends on a functional mitotic spindle checkpoint as well as on intact microtubule pulling forces. This indicates that the underlying mechanism involves cohesion fatigue in response to mitotic delay, leading to spindle checkpoint re-activation and lethal mitotic arrest. Our results point to APC/C inhibitors as promising therapeutic agents targeting cohesion-defective cancers.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/physiology , Sister Chromatid Exchange/physiology , Cell Cycle Checkpoints , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Chromosome Segregation , Humans , Mitosis/physiology , Morpholines/pharmacology , Nocodazole/pharmacology , Paclitaxel/pharmacology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sister Chromatid Exchange/drug effects , Tubulin Modulators/pharmacology
18.
Nat Commun ; 6: 8829, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26681308

ABSTRACT

Fanconi anaemia (FA) is a hereditary disease featuring hypersensitivity to DNA cross-linker-induced chromosomal instability in association with developmental abnormalities, bone marrow failure and a strong predisposition to cancer. A total of 17 FA disease genes have been reported, all of which act in a recessive mode of inheritance. Here we report on a de novo g.41022153G>A; p.Ala293Thr (NM_002875) missense mutation in one allele of the homologous recombination DNA repair gene RAD51 in an FA-like patient. This heterozygous mutation causes a novel FA subtype, 'FA-R', which appears to be the first subtype of FA caused by a dominant-negative mutation. The patient, who features microcephaly and mental retardation, has reached adulthood without the typical bone marrow failure and paediatric cancers. Together with the recent reports on RAD51-associated congenital mirror movement disorders, our results point to an important role for RAD51-mediated homologous recombination in neurodevelopment, in addition to DNA repair and cancer susceptibility.


Subject(s)
DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Fanconi Anemia/enzymology , Mutation, Missense , Acid Anhydride Hydrolases , Base Sequence , DNA Damage , DNA Repair , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Fanconi Anemia/genetics , Humans , Male , Molecular Sequence Data , Recombination, Genetic , Young Adult
19.
Anemia ; 2012: 238731, 2012.
Article in English | MEDLINE | ID: mdl-22693659

ABSTRACT

Fanconi anemia (FA) is a rare inherited syndrome with diverse clinical symptoms including developmental defects, short stature, bone marrow failure, and a high risk of malignancies. Fifteen genetic subtypes have been distinguished so far. The mode of inheritance for all subtypes is autosomal recessive, except for FA-B, which is X-linked. Cells derived from FA patients are-by definition-hypersensitive to DNA cross-linking agents, such as mitomycin C, diepoxybutane, or cisplatinum, which becomes manifest as excessive growth inhibition, cell cycle arrest, and chromosomal breakage upon cellular exposure to these drugs. Here we provide a detailed laboratory protocol for the accurate assessment of the FA diagnosis as based on mitomycin C-induced chromosomal breakage analysis in whole-blood cultures. The method also enables a quantitative estimate of the degree of mosaicism in the lymphocyte compartment of the patient.

20.
DNA Repair (Amst) ; 10(12): 1252-61, 2011 Dec 10.
Article in English | MEDLINE | ID: mdl-22036606

ABSTRACT

Fanconi anemia (FA) is a heritable disease characterized by bone marrow failure, congenital abnormalities, and cancer predisposition. The 15 identified FA genes operate in a molecular pathway to preserve genomic integrity. Within this pathway the FA core complex operates as an ubiquitin ligase that activates the complex of FANCD2 and FANCI to coordinate DNA repair. The FA core complex is formed by at least 12 proteins. However, only the FANCL subunit displays ubiquitin ligase activity. FANCA and FANCG are members of the FA core complex for which no other functions have been described than to participate in protein interactions. In this study we generated mice with combined null alleles for Fanca and Fancg to identify extended functions for these genes by characterizing the double mutant mice and cells. Double mutant a(-/-)/g(-/-) mice were born at near Mendelian frequencies without apparent developmental abnormalities. Histological analysis of a(-/-)/g(-/-) mice revealed a Leydig cell hyperplasia and frequent vacuolization of Sertoli cells in testes, while ovaries were depleted from developing follicles and displayed an interstitial cell hyperplasia. These gonadal aberrations were associated with a compromised fertility of a(-/-)/g(-/-) males and females. During the first year of life a(-/-)/g(-/-) did not develop malignancies or bone marrow failure. At the cellular level a(-/-)/g(-/-), Fanca(-/-), and Fancg(-/-) cells proved equally compromised in DNA crosslink and homology-directed repair. Overall the phenotype of a(-/-)/g(-/-) double knockout mice and cells appeared highly similar to the phenotype of Fanca or Fancg single knockouts. The lack of an augmented phenotype suggest that null mutations in Fanca or Fancg are fully epistatic, making additional important functions outside of the FA core complex highly unlikely.


Subject(s)
Epistasis, Genetic/genetics , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia/genetics , Mutation/genetics , Active Transport, Cell Nucleus/drug effects , Animals , Bone Marrow Cells/cytology , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chromosome Breakage/drug effects , Embryo, Mammalian , Female , Fertility/genetics , Fibroblasts/cytology , Fluorobenzenes/pharmacology , Hematologic Tests , Humans , Male , Mice , Ovary/metabolism , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Stem Cells/drug effects , Stem Cells/metabolism , Testis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL