Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Environ Res ; 241: 117619, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-37952855

ABSTRACT

Multi-walled carbon nanotube (MWCNT) exposure was observed to cause damages on the viability of ocular cells, however, the underlying mechanisms remain not well understood. Epigenetic alterations that regulate gene expression have been identified as a major responsiveness to environmental challenge. Thus, the aim of this study was to screen methylation-regulated genes involved in MWCNT exposure. The Illumina Human Methylation 850 K array was employed to determine the genome-wide DNA methylation profile of human retinal pigment epithelial cell line (ARPE-19) exposed to 50% inhibition concentration of MWCNTs (100 µg/ml) for 24 h or without (n = 3 for each group). Then, the transcriptome data obtained by high-throughput RNA sequencing previously were integrated with DNA methylome to identify the overlapped genes. As a result, the integrative bioinformatics analysis identified that compared with controls, FA complementation group C (FANCC) was hypermethylated and downregulated in MWCNT-exposed ARPE-19 cells. Quantitative real-time polymerase chain reaction analysis confirmed the mRNA expression level of FANCC was significantly decreased following MWCNT treatment and the addition of DNA methylation inhibitor 5-Aza-deoxycytidine (10 µM) reversed this decrease. Pyrosequencing analysis further validated the hypermethylation status at the 5'-untranslated promoter region of FANCC (cg14583550) in MWCNT-exposed ARPE-19 cells. Protein-protein interaction network and function analyses predicted that FANCC may contribute to MWCNT-induced cytotoxicity by interacting with heat shock protein 90 beta family member 1 and then upregulating cytokine interleukin-6 and apoptosis biomarker caspase 3. In conclusion, the present study links the epigenetic modification of FANCC with the pathogenesis of MWCNT-induced retinal toxicity.


Subject(s)
DNA Methylation , Nanotubes, Carbon , Humans , Nanotubes, Carbon/toxicity , Epithelial Cells , Epigenesis, Genetic , Cell Line , Fanconi Anemia Complementation Group C Protein/metabolism
2.
Nucleic Acids Res ; 51(6): 2516-2528, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36652992

ABSTRACT

At meiosis, programmed meiotic DNA double-strand breaks are repaired via homologous recombination, resulting in crossovers (COs). From a large excess of DNA double-strand breaks that are formed, only a small proportion gets converted into COs because of active mechanisms that restrict CO formation. The Fanconi anemia (FA) complex proteins AtFANCM, MHF1 and MHF2 were previously identified in a genetic screen as anti-CO factors that function during meiosis in Arabidopsis thaliana. Here, pursuing the same screen, we identify FANCC as a new anti-CO gene. FANCC was previously only identified in mammals because of low primary sequence conservation. We show that FANCC, and its physical interaction with FANCE-FANCF, is conserved from vertebrates to plants. Further, we show that FANCC, together with its subcomplex partners FANCE and FANCF, regulates meiotic recombination. Mutations of any of these three genes partially rescues CO-defective mutants, which is particularly marked in female meiosis. Functional loss of FANCC, FANCE, or FANCF results in synthetic meiotic catastrophe with the pro-CO factor MUS81. This work reveals that FANCC is conserved outside mammals and has an anti-CO role during meiosis together with FANCE and FANCF.


The Fanconi Anemia (FA) pathway is the subject of intense interest owing to the role of FA as a tumor suppressor. Three FA complex proteins, FANCM, MHF1 and MHF2, were identified as factors that suppress crossover during meiosis in the model plant Arabidopsis thaliana. Here, the authors extended these findings and identified a novel anti-crossover factor and showed that it encodes the plant FANCC homolog, which was previously thought to be vertebrate-specific. They further showed that FANCC regulates meiotic crossover together with two other FA proteins, FANCE and FANCF. This suggests that the FANCC­E­F subcomplex was already regulating DNA repair in the common ancestor of all living eukaryotes.


Subject(s)
Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group F Protein , Fanconi Anemia Complementation Group Proteins , Meiosis , Humans , Arabidopsis/genetics , Arabidopsis/metabolism , DNA/metabolism , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group F Protein/genetics , Fanconi Anemia Complementation Group F Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Homologous Recombination
3.
Nat Struct Mol Biol ; 28(3): 300-309, 2021 03.
Article in English | MEDLINE | ID: mdl-33686268

ABSTRACT

The Fanconi anemia (FA) pathway is essential for the repair of DNA interstrand crosslinks. Central to the pathway is the FA core complex, a ubiquitin ligase of nine subunits that monoubiquitinates the FANCI-FANCD2 (ID) DNA clamp. The 3.1 Å structure of the 1.1-MDa human FA core complex, described here, reveals an asymmetric assembly with two copies of all but the FANCC, FANCE and FANCF subunits. The asymmetry is crucial, as it prevents the binding of a second FANCC-FANCE-FANCF subcomplex that inhibits the recruitment of the UBE2T ubiquitin conjugating enzyme, and instead creates an ID binding site. A single active site then ubiquitinates FANCD2 and FANCI sequentially. We also present the 4.2-Å structures of the human core-UBE2T-ID-DNA complex in three conformations captured during monoubiquitination. They reveal the core-UBE2T complex remodeling the ID-DNA complex, closing the clamp on the DNA before ubiquitination. Monoubiquitination then prevents clamp opening after release from the core.


Subject(s)
DNA/metabolism , Fanconi Anemia Complementation Group Proteins/chemistry , Fanconi Anemia Complementation Group Proteins/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Binding Sites , Cryoelectron Microscopy , DNA/chemistry , DNA/ultrastructure , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group E Protein/metabolism , Fanconi Anemia Complementation Group F Protein/metabolism , Fanconi Anemia Complementation Group Proteins/ultrastructure , HEK293 Cells , Humans , Models, Molecular , Multienzyme Complexes/ultrastructure , Reproducibility of Results , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/ultrastructure , Ubiquitination , Ubiquitins/metabolism
4.
Sci Rep ; 9(1): 12524, 2019 08 29.
Article in English | MEDLINE | ID: mdl-31467304

ABSTRACT

Fanconi anemia (FA) is a genetically heterogeneous disorder with 22 disease-causing genes reported to date. In some FA genes, monoallelic mutations have been found to be associated with breast cancer risk, while the risk associations of others remain unknown. The gene for FA type C, FANCC, has been proposed as a breast cancer susceptibility gene based on epidemiological and sequencing studies. We used the Oncoarray project to genotype two truncating FANCC variants (p.R185X and p.R548X) in 64,760 breast cancer cases and 49,793 controls of European descent. FANCC mutations were observed in 25 cases (14 with p.R185X, 11 with p.R548X) and 26 controls (18 with p.R185X, 8 with p.R548X). There was no evidence of an association with the risk of breast cancer, neither overall (odds ratio 0.77, 95%CI 0.44-1.33, p = 0.4) nor by histology, hormone receptor status, age or family history. We conclude that the breast cancer risk association of these two FANCC variants, if any, is much smaller than for BRCA1, BRCA2 or PALB2 mutations. If this applies to all truncating variants in FANCC it would suggest there are differences between FA genes in their roles on breast cancer risk and demonstrates the merit of large consortia for clarifying risk associations of rare variants.


Subject(s)
Breast Neoplasms/genetics , Fanconi Anemia Complementation Group C Protein/genetics , Sequence Deletion , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Breast Neoplasms/metabolism , Case-Control Studies , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group C Protein/metabolism , Female , Genetic Predisposition to Disease , Genetic Variation , Humans
6.
Aging (Albany NY) ; 10(6): 1506-1522, 2018 Jun 21.
Article in English | MEDLINE | ID: mdl-29930218

ABSTRACT

Given importance of 22-Fanconi Anemia (FA) proteins together to act in a signaling pathway in preventing deleterious clinical symptoms, e.g. severe bone marrow failure, congenital defects, an early onset of aging and cancer, studies on each FA protein become increasingly attractive. However, an unbiased and systematic investigation of cellular effects resulting from each FA protein is missing. Here, we report roles of FA complementation C group protein (FANCC) in the protection from metabolic disorders. This study was prompted by the diabetes-prone feature displayed in FANCC knockout mice, which is not typically shown in patients with FA. We found that in cells expressing FANCC at different levels, there are representative alterations in metabolites associated with aging (glycine, citrulline, ornithine, L-asparagine, L-tyrosine, L-arginine, L-glutamine, L-leucine, L-isoleucine, L-valine, L-proline and L-alanine), Diabetes Mellitus (DM) (carbon monoxide, collagens, fatty acids, D-glucose, fumaric acid, 2-oxoglutaric acid, C3), inflammation (inosine, L-arginine, L-isoleucine, L-leucine, L-lysine, L-phenylalanine, hypoxanthine, L-methionine), and cancer ( L-methionine, sphingomyelin, acetyl-L-carnitine, L-aspartic acid, L-glutamic acid, niacinamide, phospho-rylethanolamine). We also found that FANCC can act in an FA-pathway-independent manner in tumor suppression. Collectively, featured-metabolic alterations are readouts of functional mechanisms underlying reduced tumorigenicity driven by FANCC, demonstrating close links among cancer, aging, inflammation and DM.


Subject(s)
Fanconi Anemia Complementation Group C Protein/metabolism , Metabolomics/methods , Animals , Biomarkers , Breast Neoplasms , Cell Line, Tumor , Cloning, Molecular , Female , Gene Expression Regulation , Humans , Principal Component Analysis
7.
Nat Commun ; 9(1): 2280, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29891926

ABSTRACT

Defects in DNA repair can cause various genetic diseases with severe pathological phenotypes. Fanconi anemia (FA) is a rare disease characterized by bone marrow failure, developmental abnormalities, and increased cancer risk that is caused by defective repair of DNA interstrand crosslinks (ICLs). Here, we identify the deubiquitylating enzyme USP48 as synthetic viable for FA-gene deficiencies by performing genome-wide loss-of-function screens across a panel of human haploid isogenic FA-defective cells (FANCA, FANCC, FANCG, FANCI, FANCD2). Thus, as compared to FA-defective cells alone, FA-deficient cells additionally lacking USP48 are less sensitive to genotoxic stress induced by ICL agents and display enhanced, BRCA1-dependent, clearance of DNA damage. Consequently, USP48 inactivation reduces chromosomal instability of FA-defective cells. Our results highlight a role for USP48 in controlling DNA repair and suggest it as a potential target that could be therapeutically exploited for FA.


Subject(s)
DNA Repair/genetics , DNA Repair/physiology , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , BRCA1 Protein/metabolism , CRISPR-Cas Systems , Cell Line , Chromosomal Instability , DNA Damage , Fanconi Anemia/therapy , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group G Protein/deficiency , Fanconi Anemia Complementation Group G Protein/genetics , Fanconi Anemia Complementation Group G Protein/metabolism , Fanconi Anemia Complementation Group Proteins/deficiency , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Gene Knockout Techniques , Genetic Therapy , Histones/metabolism , Humans , Mutation , Rad51 Recombinase/metabolism , Ubiquitin-Specific Proteases/deficiency , Ubiquitination
8.
J Immunol ; 200(6): 2129-2139, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29427417

ABSTRACT

Emergency (stress) granulopoiesis is an episodic process for the production of granulocytes in response to infectious challenge. We previously determined that Fanconi C, a component of the Fanconi DNA-repair pathway, is necessary for successful emergency granulopoiesis. Fanconi anemia results from mutation of any gene in this pathway and is characterized by bone marrow failure (BMF) in childhood and clonal progression in adolescence. Although murine Fanconi anemia models exhibit relatively normal steady-state hematopoiesis, FANCC-/- mice are unable to mount an emergency granulopoiesis response. Instead, these mice develop BMF and die during repeated unsuccessful emergency granulopoiesis attempts. In FANCC-/- mice, BMF is associated with extensive apoptosis of hematopoietic stem and progenitor cells through an undefined mechanism. In this study, we find that TP53 haploinsufficiency completely rescues emergency granulopoiesis in FANCC-/- mice and protects them from BMF during repeated emergency granulopoiesis episodes. Instead, such recurrent challenges accelerated clonal progression in FANCC-/-TP53+/- mice. In FANCC-/- mice, BMF during multiple emergency granulopoiesis attempts was associated with increased ataxia telangiectasia and Rad3-related protein (Atr) and p53 activation with each attempt. In contrast, we found progressive attenuation of expression and activity of Atr, and consequent p53 activation and apoptosis, in the bone marrow of FANCC-/-TP53+/- mice during this process. Therefore, activation of Atr-with consequent Fanconi-mediated DNA repair or p53-dependent apoptosis-is an essential component of emergency granulopoiesis and it protects the bone marrow from genotoxic stress during this process.


Subject(s)
Fanconi Anemia Complementation Group C Protein/metabolism , Granulocytes/metabolism , Haploinsufficiency/physiology , Leukopoiesis/physiology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/physiology , Ataxia Telangiectasia Mutated Proteins/metabolism , Bone Marrow/metabolism , DNA Damage/physiology , DNA Repair/physiology , Fanconi Anemia/metabolism , Mice
9.
J Biol Chem ; 293(11): 3937-3948, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29382715

ABSTRACT

Interferon consensus sequence-binding protein (Icsbp) is required for terminating emergency granulopoiesis, an episodic event responsible for granulocyte production in response to infections and a key component of the innate immune response. Icsbp inhibits the expression of Stat3 and C/ebpß, transcription factors essential for initiating and sustaining granulopoiesis, and activates transcription of Fanconi C (FANCC), a DNA repair protein. In prior studies, we noted accelerated bone marrow failure in Fancc-/- mice undergoing multiple episodes of emergency granulopoiesis, associated with apoptosis of bone marrow cells with unrepaired DNA damage. Additionally, we found increased expression of Fanconi C and F proteins during emergency granulopoiesis. These findings suggest that Icsbp protects the bone marrow from DNA damage by increasing activity of the Fanconi DNA repair pathway, but the mechanisms for FANCC activation during initiation of emergency granulopoiesis are unclear. In this study, we observed that Stat3 and C/ebpß activate FANCC transcription and contribute to DNA repair. Our findings indicate that FancC expression is increased during Stat3- and C/ebpß-induced initiation of emergency granulopoiesis by these transcription factors and is maintained through termination by Icsbp. Our work reveals that Stat3- and C/ebpß-mediated FancC expression is a critical component for initiating and sustaining key innate immune responses.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Fanconi Anemia Complementation Group C Protein/genetics , Gene Expression Regulation , Granulocytes/pathology , STAT3 Transcription Factor/metabolism , Transcription, Genetic , Animals , Apoptosis , Base Sequence , CCAAT-Enhancer-Binding Protein-beta/genetics , DNA Repair , Fanconi Anemia Complementation Group C Protein/metabolism , Granulocytes/metabolism , Hematopoiesis , Humans , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , STAT3 Transcription Factor/genetics , Sequence Homology , U937 Cells
10.
Nutrients ; 9(1)2017 Jan 13.
Article in English | MEDLINE | ID: mdl-28098768

ABSTRACT

Obesity induces gingival oxidative stress, which is involved in the progression of alveolar bone resorption. The antioxidant effect of hydrogen-rich water may attenuate gingival oxidative stress and prevent alveolar bone resorption in cases of obesity. We examined whether hydrogen-rich water could suppress gingival oxidative stress and alveolar bone resorption in obese rats fed a high-fat diet. Male Fischer 344 rats (n = 18) were divided into three groups of six rats each: a control group (fed a regular diet and drinking distilled water) and two experimental groups (fed a high-fat diet and drinking distilled water or hydrogen-rich water). The level of 8-hydroxydeoxyguanosine was determined to evaluate oxidative stress. The bone mineral density of the alveolar bone was analyzed by micro-computerized tomography. Obese rats, induced by a high-fat diet, showed a higher gingival level of 8-hydroxydeoxyguanosine and a lower level of alveolar bone density compared to the control group. Drinking hydrogen-rich water suppressed body weight gain, lowered gingival level of 8-hydroxydeoxyguanosine, and reduced alveolar bone resorption in rats on a high-fat diet. The results indicate that hydrogen-rich water could suppress gingival oxidative stress and alveolar bone resorption by limiting obesity.


Subject(s)
Alveolar Bone Loss/prevention & control , Diet, High-Fat/adverse effects , Drinking Water/administration & dosage , Gingiva/drug effects , Hydrogen/administration & dosage , Oxidative Stress/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Animals , Antioxidants/administration & dosage , Antioxidants/analysis , Body Weight , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/blood , Drinking Water/analysis , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/metabolism , Gene Expression , Gingiva/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Male , Rats , Rats, Inbred F344 , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Triglycerides/blood
11.
Mol Cell ; 65(2): 247-259, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-27986371

ABSTRACT

Monoubiquitination and deubiquitination of FANCD2:FANCI heterodimer is central to DNA repair in a pathway that is defective in the cancer predisposition syndrome Fanconi anemia (FA). The "FA core complex" contains the RING-E3 ligase FANCL and seven other essential proteins that are mutated in various FA subtypes. Here, we purified recombinant FA core complex to reveal the function of these other proteins. The complex contains two spatially separate FANCL molecules that are dimerized by FANCB and FAAP100. FANCC and FANCE act as substrate receptors and restrict monoubiquitination to the FANCD2:FANCI heterodimer in only a DNA-bound form. FANCA and FANCG are dispensable for maximal in vitro ubiquitination. Finally, we show that the reversal of this reaction by the USP1:UAF1 deubiquitinase only occurs when DNA is disengaged. Our work reveals the mechanistic basis for temporal and spatial control of FANCD2:FANCI monoubiquitination that is critical for chemotherapy responses and prevention of Fanconi anemia.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia/metabolism , Ubiquitination , Cell Line , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/metabolism , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group E Protein/metabolism , Fanconi Anemia Complementation Group G Protein/metabolism , Fanconi Anemia Complementation Group L Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Humans , Inhibitor of Differentiation Protein 2/metabolism , Multiprotein Complexes , Nuclear Proteins/metabolism , Protein Binding , Protein Multimerization , Recombinant Proteins/metabolism , Substrate Specificity , Time Factors , Transfection , Ubiquitin-Specific Proteases/metabolism
12.
Cell ; 165(4): 867-81, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27133164

ABSTRACT

Fanconi anemia (FA) pathway genes are important tumor suppressors whose best-characterized function is repair of damaged nuclear DNA. Here, we describe an essential role for FA genes in two forms of selective autophagy. Genetic deletion of Fancc blocks the autophagic clearance of viruses (virophagy) and increases susceptibility to lethal viral encephalitis. Fanconi anemia complementation group C (FANCC) protein interacts with Parkin, is required in vitro and in vivo for clearance of damaged mitochondria, and decreases mitochondrial reactive oxygen species (ROS) production and inflammasome activation. The mitophagy function of FANCC is genetically distinct from its role in genomic DNA damage repair. Moreover, additional genes in the FA pathway, including FANCA, FANCF, FANCL, FANCD2, BRCA1, and BRCA2, are required for mitophagy. Thus, members of the FA pathway represent a previously undescribed class of selective autophagy genes that function in immunity and organellar homeostasis. These findings have implications for understanding the pathogenesis of FA and cancers associated with mutations in FA genes.


Subject(s)
Fanconi Anemia Complementation Group C Protein/metabolism , Animals , Autophagy , Embryo, Mammalian/cytology , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Fibroblasts/metabolism , HeLa Cells , Herpesvirus 1, Human/metabolism , Humans , Inflammasomes/metabolism , Mice , Mitophagy , Reactive Oxygen Species/metabolism , Sindbis Virus/metabolism
13.
PLoS One ; 10(10): e0140612, 2015.
Article in English | MEDLINE | ID: mdl-26466335

ABSTRACT

The Fanconi anemia (FA) proteins are involved in a signaling network that assures the safeguard of chromosomes. To understand the function of FA proteins in cellular division events, we investigated the interaction between Stathmin-1 (STMN1) and the FA group C (FANCC) protein. STMN1 is a ubiquitous cytosolic protein that regulates microtubule dynamics. STMN1 activities are regulated through phosphorylation-dephosphorylation mechanisms that control assembly of the mitotic spindle, and dysregulation of STMN1 phosphorylation is associated with mitotic aberrancies leading to chromosome instability and cancer progression. Using different biochemical approaches, we showed that FANCC interacts and co-localizes with STMN1 at centrosomes during mitosis. We also showed that FANCC is required for STMN1 phosphorylation, as mutations in FANCC reduced serine 16- and 38-phosphorylated forms of STMN1. Phosphorylation of STMN1 at serine 16 is likely an event dependent on a functional FA pathway, as it is reduced in FANCA- and FANCD2-mutant cells. Furthermore, FA-mutant cells exhibited mitotic spindle anomalies such as supernumerary centrosomes and shorter mitotic spindles. These results suggest that FA proteins participate in the regulation of cellular division via the microtubule-associated protein STMN1.


Subject(s)
Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia/metabolism , Stathmin/metabolism , Cell Line , Centrosome/metabolism , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group C Protein/genetics , Humans , Mitosis , Models, Biological , Mutation , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Transport , Signal Transduction , Spindle Apparatus/metabolism , Two-Hybrid System Techniques
14.
Mol Cell Biol ; 35(22): 3829-40, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26324327

ABSTRACT

The Fanconi anemia DNA repair pathway is pivotal for the efficient repair of DNA interstrand cross-links. Here, we show that FA-defective (Fancc(-)) DT40 cells arrest in G2 phase following cross-link damage and trigger apoptosis. Strikingly, cell death was reduced in Fancc(-) cells by additional deletion of the BRCA1 tumor suppressor, resulting in elevated clonogenic survival. Increased resistance to cross-link damage was not due to loss of toxic BRCA1-mediated homologous recombination but rather through the loss of a G2 checkpoint. This proapoptotic role also required the BRCA1-A complex member ABRAXAS (FAM175A). Finally, we show that BRCA1 promotes G2 arrest and cell death by prolonging phosphorylation of Chk1 on serine 345 after DNA damage to sustain arrest. Our data imply that DNA-induced cross-link death in cells defective in the FA pathway is dependent on the ability of BRCA1 to prolong cell cycle arrest in G2 phase.


Subject(s)
Avian Proteins/metabolism , BRCA1 Protein/metabolism , DNA Repair , G2 Phase Cell Cycle Checkpoints , Protein Kinases/metabolism , Animals , Apoptosis , Avian Proteins/genetics , BRCA1 Protein/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Checkpoint Kinase 1 , Chickens , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/metabolism , Gene Deletion , Phosphorylation
15.
J Mol Cell Cardiol ; 85: 207-14, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26073630

ABSTRACT

Genome-wide association studies (GWAS) have identified genetic variants in a number of chromosomal regions that are associated with atrial fibrillation (AF). The mechanisms underlying these associations are unknown, but are likely to involve effects of the risk haplotypes on expression of neighbouring genes. To investigate the association between genetic variants at AF-associated loci and expression of nearby candidate genes in human atrial tissue and peripheral blood. Right atrial appendage (RAA) samples were collected from 122 patients undergoing cardiac surgery, of these, 12 patients also had left atrial appendage samples taken. 22 patients had a history of AF. Peripheral blood samples were collected from 405 patients undergoing diagnostic cardiac catheterisation. In order to tag genetic variation at each of nine loci, a total of 367 single nucleotide polymorphisms (SNPs) were genotyped using the Sequenom platform. Total expression of 16 candidate genes in the nine AF-associated regions was measured by quantitative PCR. The relative expression of each allele of the candidate genes was measured on the Sequenom platform using one or more transcribed SNPs to distinguish between alleles in heterozygotes. We tested association between the SNPs of interest and gene expression using total gene expression (integrating cis and trans acting sources of variation), and allelic expression ratios (specific for cis acting influences), in atrial tissue and peripheral blood. We adjusted for multiple comparisons using a Bonferroni approach. In subsidiary analyses, we compared the expression of candidate genes between patients with and without a history of AF. Total expression of 15 transcripts of 14 genes and allelic expression ratio of 14 transcripts of 14 genes in genomic regions associated with AF were measured in right atrial appendage tissue. 8 of these transcripts were also expressed in peripheral blood. Risk alleles at AF-associated SNPs were associated in cis with an increased expression of PITX2a (2.01-fold, p=6.5×10(-4)); and with decreased expression of MYOZ1 (0.39 fold; p=5.5×10(-15)), CAV1 (0.89 fold; p=5.9×10(-8)), C9orf3 (0.91 fold; 1.5×10(-5)), and FANCC (0.94-fold; p=8.9×10(-8)) in right atrial appendage. Of these five genes, only CAV1 was expressed in peripheral blood; association between the same AF risk alleles and lower expression of CAV1 was confirmed (0.91 fold decrease; p=4.2×10(-5)). A history of AF was also associated with a decrease in expression of CAV1 in both right and left atria (0.84 and 0.85 fold, respectively; p=0.03), congruent with the magnitude of the effect of the risk SNP on expression, and independent of genotype. The analyses in peripheral blood showed association between AF risk SNPs and decreased expression of KCNN3 (0.85-fold; p=2.1×10(-4)); and increased expression of SYNE2 (1.12-fold; p=7.5×10(-24)); however, these associations were not detectable in atrial tissue. We identified novel cis-acting associations in atrial tissue between AF risk SNPs and increased expression of PITX2a/b; and decreased expression of CAV1 (an association also seen in peripheral blood), C9orf3 and FANCC. We also confirmed a previously described association between AF risk variants and MYOZ1 expression. Analyses of peripheral blood illustrated tissue-specificity of cardiac eQTLs and highlight the need for larger-scale genome-wide eQTL studies in cardiac tissue. Our results suggest novel aetiological roles for genes in four AF-associated genomic regions.


Subject(s)
Aminopeptidases/metabolism , Atrial Fibrillation/genetics , Carrier Proteins/metabolism , Caveolin 1/metabolism , Fanconi Anemia Complementation Group C Protein/metabolism , Homeodomain Proteins/metabolism , Muscle Proteins/metabolism , Transcription Factors/metabolism , Aminopeptidases/genetics , Atrial Fibrillation/metabolism , Carrier Proteins/genetics , Caveolin 1/genetics , Fanconi Anemia Complementation Group C Protein/genetics , Gene Expression , Gene Expression Regulation , Genetic Predisposition to Disease , Genome-Wide Association Study , Heart Atria/metabolism , Homeodomain Proteins/genetics , Humans , Muscle Proteins/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Risk Factors , Transcription Factors/genetics , Homeobox Protein PITX2
16.
Hum Gene Ther ; 26(2): 114-26, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25545896

ABSTRACT

Genome engineering with designer nucleases is a rapidly progressing field, and the ability to correct human gene mutations in situ is highly desirable. We employed fibroblasts derived from a patient with Fanconi anemia as a model to test the ability of the clustered regularly interspaced short palindromic repeats/Cas9 nuclease system to mediate gene correction. We show that the Cas9 nuclease and nickase each resulted in gene correction, but the nickase, because of its ability to preferentially mediate homology-directed repair, resulted in a higher frequency of corrected clonal isolates. To assess the off-target effects, we used both a predictive software platform to identify intragenic sequences of homology as well as a genome-wide screen utilizing linear amplification-mediated PCR. We observed no off-target activity and show RNA-guided endonuclease candidate sites that do not possess low sequence complexity function in a highly specific manner. Collectively, we provide proof of principle for precision genome editing in Fanconi anemia, a DNA repair-deficient human disorder.


Subject(s)
Bacterial Proteins/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Deoxyribonuclease I/metabolism , Fanconi Anemia Complementation Group C Protein/metabolism , Fibroblasts/metabolism , Genome, Human , Bacterial Proteins/genetics , Base Sequence , Deoxyribonuclease I/genetics , Electroporation , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia/pathology , Fanconi Anemia/therapy , Fanconi Anemia Complementation Group C Protein/genetics , Fibroblasts/pathology , Gene Expression , Genetic Engineering , Genetic Loci , Humans , Lipids , Molecular Sequence Data , Molecular Targeted Therapy , Plasmids/chemistry , Plasmids/metabolism , Polymerase Chain Reaction/methods , Primary Cell Culture , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Streptococcus pyogenes/chemistry , Streptococcus pyogenes/enzymology , Transfection
17.
Exp Oncol ; 36(3): 212-4, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25265357

ABSTRACT

AIM: Fanconi anemia is a rare genetic disorder with high propensity for development of cancers, such as aplastic anemia, leukemia and head and neck cancers. Collagen digesting matrix metalloproteinase (MMP) enzymes have been implicated in for their role in various malignancies and to promote metastasis. Biological agents that prevent extracellular matrix digestion by the MMPs have been shown to be promising therapeutic approaches to cancer. In this study, we investigated effects of a nutrient mixture (NM) containing, ascorbic acid, lysine, proline and green tea extract, on human FANCA and FANCC lymphoblasts for viability, MMP secretion and invasion. METHODS: Human FANCA lymphoblasts GM13022 and HCS536 were challenged with NM at concentration range within 10-1000 µg/ml. Cell toxicity was assessed by Trypan blue dye exclusion test. Invasion was evaluated through Matrigel and gelatinase zymography for MMP activity. RESULTS: NM was toxic in dose dependent mode to HCS536 cells but not to GM13022 cells. GM13022 cells but not HCS536 cells exhibited MMP-9 secretion, which was inhibited by NM. Matrigel invasion was inhibited in HCS536 cells at 100 and 500 µg/ml by 27% and 93%, respectively. In GM13022 cells, the NM showed completely blocked Matrigel invasion at 500 µg/ml. CONCLUSION: NM inhibited MMP secretion and Matrigel invasion in FANCA and inhibited invasion and induced toxicity in FANCC lymphoblasts. These results suggest that the NM may have therapeutic potential in Fanconi anemia associated neoplasia.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia/drug therapy , Fanconi Anemia/pathology , Lymphocytes/drug effects , Matrix Metalloproteinases/chemistry , Plant Extracts/pharmacology , Ascorbic Acid/administration & dosage , Blotting, Western , Fanconi Anemia/metabolism , Humans , In Vitro Techniques , Lymphocytes/metabolism , Lymphocytes/pathology , Lysine/administration & dosage , Matrix Metalloproteinases/metabolism , Proline/administration & dosage , Tea/chemistry , Tumor Cells, Cultured
18.
J Cell Sci ; 127(Pt 16): 3546-54, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25015289

ABSTRACT

The aim of this study was to identify novel substrates of the FANCcore complex, the inactivation of which leads to the genetic disorder Fanconi anemia, which is associated with bone marrow failure, developmental abnormalities and a predisposition to cancer. Eight FANC proteins participate in the nuclear FANCcore complex, which functions as an E3 ubiquitin-ligase that monoubiquitylates FANCD2 and FANCI in response to replicative stress. Here, we use mass spectrometry to compare proteins from FANCcore-complex-deficient cells to those of rescued control cells after treatment with hydroxyurea, an inducer of FANCD2 monoubiquitylation. FANCD2 and FANCI appear to be the only targets of the FANCcore complex. We identify other proteins that are post-translationally modified in a FANCA- or FANCC-dependent manner. The majority of these potential targets localize to the cell membrane. Finally, we demonstrate that (a) the chemokine receptor CXCR5 is neddylated; (b) FANCA but not FANCC appears to modulate CXCR5 neddylation through an unknown mechanism; (c) CXCR5 neddylation is involved in targeting the receptor to the cell membrane; and (d) CXCR5 neddylation stimulates cell migration and motility. Our work has uncovered a pathway involving FANCA in neddylation and cell motility.


Subject(s)
Cell Membrane/metabolism , Cell Movement , Receptors, CXCR5/metabolism , Cell Membrane/genetics , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group C Protein/metabolism , Humans , NEDD8 Protein , Protein Processing, Post-Translational , Protein Transport , Proteomics , Receptors, CXCR5/genetics , Ubiquitins/metabolism
19.
PLoS One ; 9(3): e92811, 2014.
Article in English | MEDLINE | ID: mdl-24676280

ABSTRACT

The Fanconi anemia group C protein (FANCC) is one of the several proteins that comprise the Fanconi anemia (FA) network involved in genomic surveillance. FANCC is mainly cytoplasmic and has many functions, including apoptosis suppression through caspase-mediated proteolytic processing. Here, we examined the role of FANCC proteolytic fragments by identifying their binding partners. We performed a yeast two-hybrid screen with caspase-mediated FANCC cleavage products and identified the dependence receptor uncoordinated-5A (UNC5A) protein. Here, we show that FANCC physically interacts with UNC5A, a pro-apoptotic dependence receptor. FANCC interaction occurs through the UNC5A intracellular domain, specifically via its death domain. FANCC modulates cell sensitivity to UNC5A-mediated apoptosis; we observed reduced UNC5A-mediated apoptosis in the presence of FANCC and increased apoptosis in FANCC-depleted cells. Our results show that FANCC interferes with UNC5A's functions in apoptosis and suggest that FANCC may participate in developmental processes through association with the dependence receptor UNC5A.


Subject(s)
Apoptosis , Fanconi Anemia Complementation Group C Protein/metabolism , Receptors, Cell Surface/metabolism , Carrier Proteins/metabolism , Cell Line , Cytoplasm , Humans , Models, Biological , Netrin Receptors , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping/methods , Protein Transport , Receptors, Cell Surface/chemistry , Two-Hybrid System Techniques
20.
Antioxid Redox Signal ; 21(12): 1675-92, 2014 Oct 20.
Article in English | MEDLINE | ID: mdl-24483844

ABSTRACT

AIMS: This study seeks at investigating the cause of hydrocephalus, and at identifying therapeutic targets for the prevention of hydrocephalus. RESULTS: In this study, we show that inactivation of the Foxo3a gene in two mouse models of Fanconi anemia (FA) leads to the development of hydrocephalus in late embryonic stage and after birth. More than 50% of Foxo3a(-/-) Fancc(-/-) or Foxo3a(-/-) Fancd2(-/-) mice die during embryonic development or within 6 months of life as a result of hydrocephalus characterized by cranial distortion, dilation of the ventricular system, reduced thickness of the cerebral cortex, and disorganization of the ependymal cilia and subcommissural organ. Combined deficiency of Foxo3a and Fancc or Fancd2 not only impairs the self-renewal capacity but also markedly increases the apoptosis of neural stem and progenitor cells (NSPCs), leading to defective neurogenesis. Increased accumulation of reactive oxygen species (ROS) and subsequently de-regulated mitosis and ultimately apoptosis in the neural stem or progenitor cells is identified as one of the potential mechanisms of congenital obstructive hydrocephalus. INNOVATION: The work unravels a two-tier protective mechanism for preventing oxidative stress-induced hydrocephalus. CONCLUSION: The deletion of Foxo3a in FA mice increased the accumulation of ROS and subsequently de-regulated mitosis and ultimately apoptosis in the NSPCs, leading to hydrocephalus development.


Subject(s)
Fanconi Anemia Complementation Group C Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Forkhead Transcription Factors/metabolism , Hydrocephalus/metabolism , Oxidative Stress , Animals , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group C Protein/deficiency , Fanconi Anemia Complementation Group D2 Protein/deficiency , Forkhead Box Protein O3 , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Hydrocephalus/genetics , Mice , Mice, Knockout , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...