Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Oncotarget ; 7(25): 37608-37621, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-26993771

ABSTRACT

High-risk human papillomavirus (hrHPV) types induce immortalization of primary human epithelial cells. Previously we demonstrated that immortalization of human foreskin keratinocytes (HFKs) is HPV type dependent, as reflected by the presence or absence of a crisis period before reaching immortality. This study determined how the immortalization capacity of ten hrHPV types relates to DNA damage induction and overall genomic instability in HFKs.Twenty five cell cultures obtained by transduction of ten hrHPV types (i.e. HPV16/18/31/33/35/45/51/59/66/70 E6E7) in two or three HFK donors each were studied.All hrHPV-transduced HFKs showed an increased number of double strand DNA breaks compared to controls, without exhibiting significant differences between types. However, immortal descendants of HPV-transduced HFKs that underwent a prior crisis period (HPV45/51/59/66/70-transduced HFKs) showed significantly more chromosomal aberrations compared to those without crisis (HPV16/18/31/33/35-transduced HFKs). Notably, the hTERT locus at 5p was exclusively gained in cells with a history of crisis and coincided with increased expression. Chromothripsis was detected in one cell line in which multiple rearrangements within chromosome 8 resulted in a gain of MYC.Together we demonstrated that upon HPV-induced immortalization, the number of chromosomal aberrations is inversely related to the viral immortalization capacity. We propose that hrHPV types with reduced immortalization capacity in vitro, reflected by a crisis period, require more genetic host cell aberrations to facilitate immortalization than types that can immortalize without crisis. This may in part explain the observed differences in HPV-type prevalence in cervical cancers and emphasizes that changes in the host cell genome contribute to HPV-induced carcinogenesis.


Subject(s)
Human papillomavirus 16/genetics , Human papillomavirus 18/genetics , Papillomavirus Infections/virology , Chromosomal Instability , Human papillomavirus 16/metabolism , Human papillomavirus 18/metabolism , Humans
2.
Turk J Pediatr ; 57(3): 282-5, 2015.
Article in English | MEDLINE | ID: mdl-26701949

ABSTRACT

Fanconi anemia (FA) is an inherited disorder characterized by malformations, marrow failure, and predisposition to cancer. Birth defects and laboratory features are characteristic and helpful in diagnosis, when present. Chromosome fragility is pathognomonic in the diagnosis. However, in some cases, there are no obvious physical anomalies or suggestive hematologic abnormalities, and inconclusive diagnostic tests have also been described. In such cases, a molecular diagnosis is required. This approach presents some advantages, especially in populations with a high incidence of FA and of consanguinity. Herein, we present a case with mild phenotypic features, inconclusive hematological findings and a negative breakage test. The diagnosis of FA was confirmed with next-generation sequencing. To our knowledge, this is the first publication of a FA patient being molecularly diagnosed utilizing this method since its introduction. Given its technical and financial features, we suggest that next-generation sequencing might be an alternative first-line diagnostic test for selected cases from particular populations.


Subject(s)
Fanconi Anemia/diagnosis , Fanconi Anemia/genetics , Child, Preschool , Chromosome Breakage , Diagnosis, Differential , Humans , Male , Molecular Diagnostic Techniques , Physical Examination
3.
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
4.
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
5.
Hum Mutat ; 36(5): 562-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25754594

ABSTRACT

The diagnosis of VACTERL syndrome can be elusive, especially in the prenatal life, due to the presence of malformations that overlap those present in other genetic conditions, including the Fanconi anemia (FA). We report on three VACTERL cases within two families, where the two who arrived to be born died shortly after birth due to severe organs' malformations. The suspicion of VACTERL association was based on prenatal ultrasound assessment and postnatal features. Subsequent chromosome breakage analysis suggested the diagnosis of FA. Finally, by next-generation sequencing based on the analysis of the exome in one family and of a panel of Fanconi genes in the second one, we identified novel FANCL truncating mutations in both families. We used ectopic expression of wild-type FANCL to functionally correct the cellular FA phenotype for both mutations. Our study emphasizes that the diagnosis of FA should be considered when VACTERL association is suspected. Furthermore, we show that loss-of-function mutations in FANCL result in a severe clinical phenotype characterized by early postnatal death.


Subject(s)
Anal Canal/abnormalities , Esophagus/abnormalities , Fanconi Anemia Complementation Group L Protein/genetics , Fanconi Anemia/diagnosis , Fanconi Anemia/genetics , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/genetics , Kidney/abnormalities , Limb Deformities, Congenital/diagnosis , Limb Deformities, Congenital/genetics , Mutation , Phenotype , Spine/abnormalities , Trachea/abnormalities , Abortion, Induced , Chromosome Breakage , Diagnosis, Differential , Exome , Female , High-Throughput Nucleotide Sequencing , Humans , Infant, Newborn , Live Birth , Male , Pregnancy , Prenatal Diagnosis , Severity of Illness Index
6.
DNA Repair (Amst) ; 26: 54-64, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25583207

ABSTRACT

The encouraging response rates of BRCA1- and BRCA2-mutated cancers toward PARP inhibitors make it worthwhile to identify other potential determinants of PARP inhibitor responsiveness. Since the Fanconi anemia (FA) pathway coordinates several DNA repair pathways, including homologous recombination in which BRCA1 and BRCA2 play important roles, we investigated whether this pathway harbors other predictors of PARP inhibitor sensitivity. Lymphoblastoid cell lines derived from individuals with FA or clinically related syndromes, such as Warsaw breakage syndrome, were tested for PARP inhibitor sensitivity. Remarkably, we found a strong variability in PARP inhibitor sensitivity among different FANCD1/BRCA2-deficient lymphoblasts, suggesting that PARP inhibitor response depends on the type of FANCD1/BRCA2 mutation. We identified the DNA helicases FANCM and DDX11 as determinants of PARP inhibitor response. These results may extend the utility of PARP inhibition as effective anticancer treatment.


Subject(s)
DEAD-box RNA Helicases/genetics , DNA Helicases/genetics , Enzyme Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , BRCA2 Protein/genetics , Cell Line, Transformed , Fanconi Anemia/genetics , Female , Fluorobenzenes/pharmacology , Gene Knockdown Techniques , Humans , Male , Phthalazines/pharmacology
7.
Springerplus ; 3: 381, 2014.
Article in English | MEDLINE | ID: mdl-25161863

ABSTRACT

Fanconi anemia (FA) is a genetically heterogeneous syndrome associated with increased cancer predisposition. The underlying genes govern the FA pathway which functions to protect the genome during the S-phase of the cell cycle. While upregulation of FA genes has been linked to chemotherapy resistance, little is known about their regulation in response to proliferative stimuli. The purpose of this study was to examine how FA genes are regulated, especially in relation to the cell cycle, in order to reveal their possible participation in biochemical networks. Expression of 14 FA genes was monitored in two human cell-cycle models and in two RB1/E2F pathway-associated primary cancers, retinoblastoma and basal breast cancer. In silico studies were performed to further evaluate coregulation and identify connected networks and diseases. Only FANCA was consistently induced over 2-fold; FANCF failed to exhibit any regulatory fluctuations. Two tools exploiting public data sets indicated coregulation of FANCA with BRCA1. Upregulation of FANCA and BRCA1 correlated with upregulation of E2F3. Genes coregulated with both FANCA and BRCA1 were enriched for MeSH-Term id(s) genomic instability, microcephaly, and Bloom syndrome, and enriched for the cellular component centrosome. The regulation of FA genes appears highly divergent. In RB1-linked tumors, upregulation of FA network genes was associated with reduced expression of FANCF. FANCA and BRCA1 may jointly act in a subnetwork - supporting vital function(s) at the subcellular level (centrosome) as well as at the level of embryonic development (mechanisms controlling head circumference).

8.
Clin Cancer Res ; 20(18): 4816-26, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24963051

ABSTRACT

PURPOSE: Poly(ADP-ribose) polymerase (PARP) inhibitors are promising targeted treatment options for hereditary breast tumors with a homologous recombination (HR) deficiency caused by BRCA1 or BRCA2 mutations. However, the functional consequence of BRCA gene mutations is not always known and tumors can be HR deficient for other reasons than BRCA gene mutations. Therefore, we aimed to develop a functional test to determine HR activity in tumor samples to facilitate selection of patients eligible for PARP inhibitor treatment. EXPERIMENTAL DESIGN: We obtained 54 fresh primary breast tumor samples from patients undergoing surgery. We determined their HR capacity by studying the formation of ionizing radiation induced foci (IRIF) of the HR protein RAD51 after ex vivo irradiation of these organotypic breast tumor samples. Tumors showing impaired RAD51 IRIF formation were subjected to genetic and epigenetic analysis. RESULTS: Five of 45 primary breast tumors with sufficient numbers of proliferating tumor cells were RAD51 IRIF formation deficient (11%, 95% CI, 5%-24%). This HR defect was significantly associated with triple-negative breast cancer (OR, 57; 95% CI, 3.9-825; P = 0.003). Two of five HR-deficient tumors were not caused by mutations in the BRCA genes, but by BRCA1 promoter hypermethylation. CONCLUSION: The functional RAD51 IRIF assay faithfully identifies HR-deficient tumors and has clear advantages over gene sequencing. It is a relatively easy assay that can be performed on biopsy material, making it a powerful tool to select patients with an HR-deficient cancer for PARP inhibitor treatment in the clinic.


Subject(s)
Biological Assay/methods , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Homologous Recombination/radiation effects , Rad51 Recombinase/radiation effects , Animals , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Female , Fluorescent Antibody Technique , Genes, BRCA1 , Genes, BRCA2 , Humans , Mice , Poly(ADP-ribose) Polymerase Inhibitors , Xenograft Model Antitumor Assays
9.
Hum Mutat ; 35(4): 442-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24395671

ABSTRACT

Fanconi anemia (FA) is a rare recessive disorder with chromosomal instability, congenital abnormalities, and a high cancer risk. The breast cancer susceptibility gene BRCA2 (FANCD1) is one of the 16 genes involved in this recessive disease. We have identified a novel mutation of the splice donor site of intron 1 in the noncoding region of BRCA2 in a Japanese FA family. This mutation may account for the FA phenotype in a patient originally reported to have biallelic mutations in BRCA2. Subsequent functional studies revealed that one of the mutations, K2729N, was a neutral change. As reported here, a more careful analysis resulted in the identification of a novel splice site mutation. Functional analysis using a mouse embryonic stem cell-based assay revealed that it causes aberrant splicing, reduced transcript levels and hypersensitivity to DNA damaging agents, suggesting that it is likely to be pathogenic. Although similar pathogenic variants in the noncoding region of BRCA1 and 2 were not identified in a cohort of 752 familial breast cancer cases, we still think this finding is relevant for mutation analysis in Hereditary Breast and Ovarian Cancer Syndrome families in a diagnostic setting.


Subject(s)
BRCA2 Protein/genetics , Fanconi Anemia/diagnosis , Fanconi Anemia/genetics , Hereditary Breast and Ovarian Cancer Syndrome/diagnosis , Hereditary Breast and Ovarian Cancer Syndrome/genetics , Animals , BRCA1 Protein/genetics , Base Sequence , Cells, Cultured , DNA Mutational Analysis , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Introns , Mice , Molecular Sequence Data , Mutation, Missense , Pedigree , RNA Splice Sites
10.
Mol Cell ; 52(3): 434-46, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24207054

ABSTRACT

The replicative machinery encounters many impediments, some of which can be overcome by lesion bypass or replication restart pathways, leaving repair for a later time. However, interstrand crosslinks (ICLs), which preclude DNA unwinding, are considered absolute blocks to replication. Current models suggest that fork collisions, either from one or both sides of an ICL, initiate repair processes required for resumption of replication. To test these proposals, we developed a single-molecule technique for visualizing encounters of replication forks with ICLs as they occur in living cells. Surprisingly, the most frequent patterns were consistent with replication traverse of an ICL, without lesion repair. The traverse frequency was strongly reduced by inactivation of the translocase and DNA binding activities of the FANCM/MHF complex. The results indicate that translocase-based mechanisms enable DNA synthesis to continue past ICLs and that these lesions are not always absolute blocks to replication.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , DNA Helicases/metabolism , DNA Replication/genetics , DNA/biosynthesis , Fanconi Anemia/genetics , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cricetinae , DNA Helicases/genetics , DNA-Binding Proteins , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Humans , Mice , Multiprotein Complexes/metabolism , Tumor Suppressor Proteins/genetics
11.
PLoS One ; 8(4): e62017, 2013.
Article in English | MEDLINE | ID: mdl-23620800

ABSTRACT

Fanconi anemia (FA) is a heterogeneous recessive disorder associated with a markedly elevated risk to develop cancer. To date sixteen FA genes have been identified, three of which predispose heterozygous mutation carriers to breast cancer. The FA proteins work together in a genome maintenance pathway, the so-called FA/BRCA pathway which is important during the S phase of the cell cycle. Since not all FA patients can be linked to (one of) the sixteen known complementation groups, new FA genes remain to be identified. In addition the complex FA network remains to be further unravelled. One of the FA genes, FANCI, has been identified via a combination of bioinformatic techniques exploiting FA protein properties and genetic linkage. The aim of this study was to develop a prioritization approach for proteins of the entire human proteome that potentially interact with the FA/BRCA pathway or are novel candidate FA genes. To this end, we combined the original bioinformatics approach based on the properties of the first thirteen FA proteins identified with publicly available tools for protein-protein interactions, literature mining (Nermal) and a protein function prediction tool (FuncNet). Importantly, the three newest FA proteins FANCO/RAD51C, FANCP/SLX4, and XRCC2 displayed scores in the range of the already known FA proteins. Likewise, a prime candidate FA gene based on next generation sequencing and having a very low score was subsequently disproven by functional studies for the FA phenotype. Furthermore, the approach strongly enriches for GO terms such as DNA repair, response to DNA damage stimulus, and cell cycle-regulated genes. Additionally, overlaying the top 150 with a haploinsufficiency probability score, renders the approach more tailored for identifying breast cancer related genes. This approach may be useful for prioritization of putative novel FA or breast cancer genes from next generation sequencing efforts.


Subject(s)
Computational Biology/methods , Fanconi Anemia Complementation Group Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Cell Cycle/genetics , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Haploinsufficiency/genetics , Humans , Molecular Sequence Annotation , Proteome , Reproducibility of Results
12.
Am J Hum Genet ; 92(5): 800-6, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23623386

ABSTRACT

Fanconi anemia (FA) is a rare genomic instability disorder characterized by progressive bone marrow failure and predisposition to cancer. FA-associated gene products are involved in the repair of DNA interstrand crosslinks (ICLs). Fifteen FA-associated genes have been identified, but the genetic basis in some individuals still remains unresolved. Here, we used whole-exome and Sanger sequencing on DNA of unclassified FA individuals and discovered biallelic germline mutations in ERCC4 (XPF), a structure-specific nuclease-encoding gene previously connected to xeroderma pigmentosum and segmental XFE progeroid syndrome. Genetic reversion and wild-type ERCC4 cDNA complemented the phenotype of the FA cell lines, providing genetic evidence that mutations in ERCC4 cause this FA subtype. Further biochemical and functional analysis demonstrated that the identified FA-causing ERCC4 mutations strongly disrupt the function of XPF in DNA ICL repair without severely compromising nucleotide excision repair. Our data show that depending on the type of ERCC4 mutation and the resulting balance between both DNA repair activities, individuals present with one of the three clinically distinct disorders, highlighting the multifunctional nature of the XPF endonuclease in genome stability and human disease.


Subject(s)
DNA-Binding Proteins/genetics , Deoxyribonucleases/genetics , Fanconi Anemia/genetics , Genetic Predisposition to Disease/genetics , Phenotype , Apoptosis/genetics , Apoptosis/radiation effects , Base Sequence , Exome/genetics , Fanconi Anemia/pathology , Germ-Line Mutation/genetics , Humans , Immunoblotting , Immunoprecipitation , Molecular Sequence Data , Sequence Analysis, DNA , Ultraviolet Rays
13.
Dis Model Mech ; 6(1): 40-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23268537

ABSTRACT

Fanconi anaemia (FA) is a rare autosomal recessive or X-linked inherited disease characterised by an increased incidence of bone marrow failure (BMF), haematological malignancies and solid tumours. Cells from individuals with FA show a pronounced sensitivity to DNA interstrand crosslink (ICL)-inducing agents, which manifests as G2-M arrest, chromosomal aberrations and reduced cellular survival. To date, mutations in at least 15 different genes have been identified that cause FA; the products of all of these genes are thought to function together in the FA pathway, which is essential for ICL repair. Rapidly following the discovery of FA genes, mutant mice were generated to study the disease and the affected pathway. These mutant mice all show the characteristic cellular ICL-inducing agent sensitivity, but only partially recapitulate the developmental abnormalities, anaemia and cancer predisposition seen in individuals with FA. Therefore, the usefulness of modelling FA in mice has been questioned. In this Review, we argue that such scepticism is unjustified. We outline that haematopoietic defects and cancer predisposition are manifestations of FA gene defects in mice, albeit only in certain genetic backgrounds and under certain conditions. Most importantly, recent work has shown that developmental defects in FA mice also arise with concomitant inactivation of acetaldehyde metabolism, giving a strong clue about the nature of the endogenous lesion that must be repaired by the functional FA pathway. This body of work provides an excellent example of a paradox in FA research: that the dissimilarity, rather than the similarity, between mice and humans can provide insight into human disease. We expect that further study of mouse models of FA will help to uncover the mechanistic background of FA, ultimately leading to better treatment options for the disease.


Subject(s)
Fanconi Anemia/genetics , Animals , Congenital Abnormalities/genetics , Disease Models, Animal , Fanconi Anemia/etiology , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Genes, Lethal , Humans , Mice , Mice, Mutant Strains , Mutation , Neoplasms/genetics , Species Specificity , Telomere/genetics
14.
Hum Mutat ; 34(1): 70-3, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22911665

ABSTRACT

SLX4/FANCP is a recently discovered novel disease gene for Fanconi anemia (FA), a rare recessive disorder characterized by chromosomal instability and increased cancer susceptibility. Three of the 15 FA genes are breast cancer susceptibility genes in heterozygous mutation carriers--BRCA2, PALB2, and BRIP1. To investigate if defects in SLX4 also predispose to breast cancer, the gene was sequenced in a cohort of 729 BRCA1/BRCA2-negative familial breast cancer cases. We identified a single splice site mutation (c.2013+2T>A), which causes a frameshift by skipping of exon 8. We also identified 39 missense variants, four of which were selected for functional testing in a Mitomycin C-induced growth inhibition assay, and appeared indistinguishable from wild type. Although this is the first study that describes a truncating SLX4 mutation in breast cancer patients, our data indicate that germline mutations in SLX4 are very rare and are unlikely to make a significant contribution to familial breast cancer.


Subject(s)
Breast Neoplasms, Male/genetics , Breast Neoplasms/genetics , Mutation , Recombinases/genetics , Adult , Aged , Aged, 80 and over , Cohort Studies , DNA Mutational Analysis , Family Health , Fanconi Anemia/genetics , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Mutation, Missense , RNA Splice Sites/genetics
15.
Hum Mutat ; 34(1): 93-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23033263

ABSTRACT

Fanconi anemia (FA) is a rare genetic disorder characterized by congenital malformations, progressive bone marrow failure (BMF), and susceptibility to malignancies. FA is caused by biallelic or hemizygous mutations in one of 15 known FA genes, whose products are involved in the FA/BRCA DNA damage response pathway. Here, we report on a patient with previously unknown mutations of the most recently identified FA gene, SLX4/FANCP. Whole exome sequencing (WES) revealed a nonsense mutation and an unusual splice site mutation resulting in the partial replacement of exonic with intronic bases, thereby removing a nuclear localization signal. Immunoblotting detected no residual SLX4 protein, which was consistent with abrogated interactions with XPF/ERCC1 and MUS81/EME1. This cellular finding did not result in a more severe clinical phenotype than that of previously reported FA-P patients. Our study additionally exemplifies the versatility of WES for the detection of mutations in heterogenic disorders such as FA.


Subject(s)
Exome/genetics , Fanconi Anemia/genetics , Mutation , Recombinases/genetics , Base Sequence , Codon, Nonsense , DNA Mutational Analysis , Fanconi Anemia/metabolism , Female , Humans , Immunoblotting , Molecular Sequence Data , RNA Splice Sites/genetics , Recombinases/metabolism , Young Adult
16.
Anemia ; 2012: 603253, 2012.
Article in English | MEDLINE | ID: mdl-22778927

ABSTRACT

Fanconi anemia (FA) is a rare inherited disease characterized by developmental defects, short stature, bone marrow failure, and a high risk of malignancies. FA is heterogeneous: 15 genetic subtypes have been distinguished so far. A clinical diagnosis of FA needs to be confirmed by testing cells for sensitivity to cross-linking agents in a chromosomal breakage test. As a second step, DNA testing can be employed to elucidate the genetic subtype of the patient and to identify the familial mutations. This knowledge allows preimplantation genetic diagnosis (PGD) and enables prenatal DNA testing in future pregnancies. Although simultaneous testing of all FA genes by next generation sequencing will be possible in the near future, this technique will not be available immediately for all laboratories. In addition, in populations with strong founder mutations, a limited test using Sanger sequencing and MLPA will be a cost-effective alternative. We describe a strategy and optimized conditions for the screening of FANCA, FANCB, FANCC, FANCE, FANCF, and FANCG and present the results obtained in a cohort of 54 patients referred to our diagnostic service since 2008. In addition, the follow up with respect to genetic counseling and carrier screening in the families is discussed.

17.
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.

18.
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
19.
Anemia ; 2012: 132856, 2012.
Article in English | MEDLINE | ID: mdl-22720145

ABSTRACT

Fanconi anemia (FA) is a rare genetic instability syndrome characterized by developmental defects, bone marrow failure, and a high cancer risk. Fifteen genetic subtypes have been distinguished. The majority of patients (≈85%) belong to the subtypes A (≈60%), C (≈15%) or G (≈10%), while a minority (≈15%) is distributed over the remaining 12 subtypes. All subtypes seem to fit within the "classical" FA phenotype, except for D1 and N patients, who have more severe clinical symptoms. Since FA patients need special clinical management, the diagnosis should be firmly established, to exclude conditions with overlapping phenotypes. A valid FA diagnosis requires the detection of pathogenic mutations in a FA gene and/or a positive result from a chromosomal breakage test. Identification of the pathogenic mutations is also important for adequate genetic counselling and to facilitate prenatal or preimplantation genetic diagnosis. Here we describe and validate a comprehensive protocol for the molecular diagnosis of FA, based on massively parallel sequencing. We used this approach to identify BRCA2, FANCD2, FANCI and FANCL mutations in novel unclassified FA patients.

20.
Cancer Discov ; 2(1): 14-5, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22585162

ABSTRACT

The genetic risk factors that contribute to pancreatic cancers are largely unknown. A new next-generation sequencing study by Roberts and colleagues now adds ATM to the list of pancreatic ductal adenocarcinoma predisposition genes.


Subject(s)
Ataxia Telangiectasia/genetics , Carcinoma, Pancreatic Ductal/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Pancreatic Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Ataxia Telangiectasia Mutated Proteins , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...