Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nucleic Acids Res ; 52(5): 2372-2388, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38214234

ABSTRACT

Pediatric high-grade gliomas (pHGG) are devastating and incurable brain tumors with recurrent mutations in histone H3.3. These mutations promote oncogenesis by dysregulating gene expression through alterations of histone modifications. We identify aberrant DNA repair as an independent mechanism, which fosters genome instability in H3.3 mutant pHGG, and opens new therapeutic options. The two most frequent H3.3 mutations in pHGG, K27M and G34R, drive aberrant repair of replication-associated damage by non-homologous end joining (NHEJ). Aberrant NHEJ is mediated by the DNA repair enzyme polynucleotide kinase 3'-phosphatase (PNKP), which shows increased association with mutant H3.3 at damaged replication forks. PNKP sustains the proliferation of cells bearing H3.3 mutations, thus conferring a molecular vulnerability, specific to mutant cells, with potential for therapeutic targeting.


Subject(s)
Brain Neoplasms , Glioma , Histones , Child , Humans , Brain Neoplasms/pathology , DNA Repair/genetics , DNA Repair Enzymes/metabolism , Glioma/pathology , Histones/genetics , Histones/metabolism , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics
2.
Mol Cell ; 57(1): 123-37, 2015 01 08.
Article in English | MEDLINE | ID: mdl-25533188

ABSTRACT

The SLX4 Fanconi anemia protein is a tumor suppressor that may act as a key regulator that engages the cell into specific genome maintenance pathways. Here, we show that the SLX4 complex is a SUMO E3 ligase that SUMOylates SLX4 itself and the XPF subunit of the DNA repair/recombination XPF-ERCC1 endonuclease. This SLX4-dependent activity is mediated by a remarkably specific interaction between SLX4 and the SUMO-charged E2 conjugating enzyme UBC9 and relies not only on newly identified SUMO-interacting motifs (SIMs) in SLX4 but also on its BTB domain. In contrast to its ubiquitin-binding UBZ4 motifs, SLX4 SIMs are dispensable for its DNA interstrand crosslink repair functions. Instead, while detrimental in response to global replication stress, the SUMO E3 ligase activity of the SLX4 complex is critical to prevent mitotic catastrophe following common fragile site expression.


Subject(s)
DNA Repair , DNA-Binding Proteins/metabolism , Genome , Protein Subunits/metabolism , Recombinases/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Cell Line, Tumor , DNA Replication , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genomic Instability , Humans , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , Protein Subunits/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinases/genetics , Sequence Alignment , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics
3.
PLoS Genet ; 13(5): e1006784, 2017 May.
Article in English | MEDLINE | ID: mdl-28505193

ABSTRACT

INT6/eIF3e is a highly conserved component of the translation initiation complex that interacts with both the 26S proteasome and the COP9 signalosome, two complexes implicated in ubiquitin-mediated protein degradation. The INT6 gene was originally identified as the insertion site of the mouse mammary tumor virus (MMTV), and later shown to be involved in human tumorigenesis. Here we show that depletion of the Drosophila orthologue of INT6 (Int6) results in short mitotic spindles and deformed centromeres and kinetochores with low intra-kinetochore distance. Poleward flux of microtubule subunits during metaphase is reduced, although fluorescence recovery after photobleaching (FRAP) demonstrates that microtubules remain dynamic both near the kinetochores and at spindle poles. Mitotic progression is delayed during metaphase due to the activity of the spindle assembly checkpoint (SAC). Interestingly, a deubiquitinated form of the kinesin Klp67A (a putative orthologue of human Kif18A) accumulates near the kinetochores in Int6-depleted cells. Consistent with this finding, Klp67A overexpression mimics the Int6 RNAi phenotype. Furthermore, simultaneous depletion of Int6 and Klp67A results in a phenotype identical to RNAi of just Klp67A, which indicates that Klp67A deficiency is epistatic over Int6 deficiency. We propose that Int6-mediated ubiquitination is required to control the activity of Klp67A. In the absence of this control, excess of Klp67A at the kinetochore suppresses microtubule plus-end polymerization, which in turn results in reduced microtubule flux, spindle shortening, and centromere/kinetochore deformation.


Subject(s)
Eukaryotic Initiation Factor-3/genetics , Kinetochores/metabolism , Microtubules/metabolism , Animals , Cell Line , Drosophila/genetics , Drosophila/metabolism , Drosophila/ultrastructure , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Eukaryotic Initiation Factor-3/metabolism , Kinetochores/ultrastructure , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/genetics , Mitosis , Ubiquitination
4.
Blood ; 124(24): 3613-23, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25261197

ABSTRACT

Fanconi anemia (FA) is an inherited chromosomal instability syndrome that is characterized by progressive bone marrow failure. One of the main causes of morbidity and mortality in FA is a bleeding tendency, resulting from low platelet counts. Platelets are the final products of megakaryocyte (MK) maturation. Here, we describe a previously unappreciated role of Fanconi anemia group A protein (Fanca) during the endomitotic process of MK differentiation. Fanca deficiency leads to the accumulation of MKs with low nuclear ploidy and to decreased platelet production. We show, for the first time, that Fanca(-/-) mice are characterized by limited number and proliferative capacity of MK progenitors. Defective megakaryopoiesis of Fanca(-/-) cells is associated with the formation of nucleoplasmic bridges and increased chromosomal instability, indicating that inaccurate endoreplication and karyokinesis occur during MK polyploidization. Sustained DNA damage forces Fanca(-/-) MKs to enter a senescence-like state. Furthermore, inhibition of the Rho-associated kinase, a regulator of cytokinesis, improves the polyploidization of Fanca(-/-) MKs but greatly increases their genomic instability and diminishes their differentiation potential, supporting the notion that accumulation of DNA damage through endomitotic cycles affects MK maturation. Our study indicates that Fanca expression during endomitosis is crucial for normal megakaryopoiesis and platelet production.


Subject(s)
Fanconi Anemia Complementation Group A Protein , Gene Expression Regulation/genetics , Megakaryocytes/metabolism , Thrombocytopenia , Thrombopoiesis/genetics , Animals , Cellular Senescence/genetics , Chromosomal Instability/genetics , Fanconi Anemia Complementation Group A Protein/biosynthesis , Fanconi Anemia Complementation Group A Protein/genetics , Megakaryocytes/pathology , Mice , Mice, Knockout , Mitosis , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
5.
Nucleic Acids Res ; 39(13): 5459-73, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21421559

ABSTRACT

Deciphering the crosstalk between a host cell and a virus during infection is important not only to better define viral biology but also to improve our understanding of cellular processes. We identified the FANC pathway as a helper of viral replication and recombination by searching for cellular targets that are modified by adenovirus (Ad) infection and are involved in its outcome. This pathway, which is involved in the DNA damage response and checkpoint control, is altered in Fanconi anaemia, a rare cancer predisposition syndrome. We show here that Ad5 infection activates the FANC pathway independent of the classical DNA damage response. Infection with a non-replicating Ad shows that the presence of viral DNA is not sufficient to induce the monoubiquitination of FANCD2 but still activates the DNA damage response coordinated by phospho-NBS1 and phospho-CHK1. E1A expression alone fails to induce FANCD2 monoubiquitination, indicating that a productive viral infection and/or replication is required for FANC pathway activation. Our data indicate that Ad5 infection induces FANCD2 activation to promote its own replication. Specifically, we show that FANCD2 is involved in the recombination process that accompanies viral DNA replication. This study provides evidence of a DNA damage-independent function of the FANC pathway and identifies a cellular system involved in Ad5 recombination.


Subject(s)
Adenoviridae/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Recombination, Genetic , Virus Replication , Adenoviridae/physiology , Adenovirus E1A Proteins/metabolism , Cell Line , Cells, Cultured , DNA Damage , DNA Replication , Fanconi Anemia Complementation Group Proteins/metabolism , Humans , Kinetics , Ubiquitination
6.
Commun Biol ; 5(1): 1395, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36543851

ABSTRACT

Replication stress (RS) is a leading cause of genome instability and cancer development. A substantial source of endogenous RS originates from the encounter between the transcription and replication machineries operating on the same DNA template. This occurs predominantly under specific contexts, such as oncogene activation, metabolic stress, or a deficiency in proteins that specifically act to prevent or resolve those transcription-replication conflicts (TRCs). One such protein is Senataxin (SETX), an RNA:DNA helicase involved in resolution of TRCs and R-loops. Here we identify a synthetic lethal interaction between SETX and proteins of the Fanconi anemia (FA) pathway. Depletion of SETX induces spontaneous under-replication and chromosome fragility due to active transcription and R-loops that persist in mitosis. These fragile loci are targeted by the Fanconi anemia protein, FANCD2, to facilitate the resolution of under-replicated DNA, thus preventing chromosome mis-segregation and allowing cells to proliferate. Mechanistically, we show that FANCD2 promotes mitotic DNA synthesis that is dependent on XPF and MUS81 endonucleases. Importantly, co-depleting FANCD2 together with SETX impairs cancer cell proliferation, without significantly affecting non-cancerous cells. Therefore, we uncovered a synthetic lethality between SETX and FA proteins for tolerance of transcription-mediated RS that may be exploited for cancer therapy.


Subject(s)
DNA Helicases , Fanconi Anemia Complementation Group D2 Protein , Neoplasms , RNA Helicases , Humans , DNA , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Replication , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Neoplasms/genetics , Neoplasms/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism
7.
PLoS Genet ; 4(7): e1000126, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18797514

ABSTRACT

RNAi screens have, to date, identified many genes required for mitotic divisions of Drosophila tissue culture cells. However, the inventory of such genes remains incomplete. We have combined the powers of bioinformatics and RNAi technology to detect novel mitotic genes. We found that Drosophila genes involved in mitosis tend to be transcriptionally co-expressed. We thus constructed a co-expression-based list of 1,000 genes that are highly enriched in mitotic functions, and we performed RNAi for each of these genes. By limiting the number of genes to be examined, we were able to perform a very detailed phenotypic analysis of RNAi cells. We examined dsRNA-treated cells for possible abnormalities in both chromosome structure and spindle organization. This analysis allowed the identification of 142 mitotic genes, which were subdivided into 18 phenoclusters. Seventy of these genes have not previously been associated with mitotic defects; 30 of them are required for spindle assembly and/or chromosome segregation, and 40 are required to prevent spontaneous chromosome breakage. We note that the latter type of genes has never been detected in previous RNAi screens in any system. Finally, we found that RNAi against genes encoding kinetochore components or highly conserved splicing factors results in identical defects in chromosome segregation, highlighting an unanticipated role of splicing factors in centromere function. These findings indicate that our co-expression-based method for the detection of mitotic functions works remarkably well. We can foresee that elaboration of co-expression lists using genes in the same phenocluster will provide many candidate genes for small-scale RNAi screens aimed at completing the inventory of mitotic proteins.


Subject(s)
Drosophila/genetics , Gene Expression , Genes, Insect , Mitosis/genetics , RNA Interference , Animals , Chromosome Segregation , Cytokinesis , Drosophila/metabolism , RNA, Double-Stranded/metabolism , Spindle Apparatus/genetics , Spindle Apparatus/metabolism
8.
Commun Biol ; 4(1): 127, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514811

ABSTRACT

Common fragile sites (CFSs) are genomic regions frequently involved in cancer-associated rearrangements. Most CFSs lie within large genes, and their instability involves transcription- and replication-dependent mechanisms. Here, we uncover a role for the mitochondrial stress response pathway in the regulation of CFS stability in human cells. We show that FANCD2, a master regulator of CFS stability, dampens the activation of the mitochondrial stress response and prevents mitochondrial dysfunction. Genetic or pharmacological activation of mitochondrial stress signaling induces CFS gene expression and concomitant relocalization to CFSs of FANCD2. FANCD2 attenuates CFS gene transcription and promotes CFS gene stability. Mechanistically, we demonstrate that the mitochondrial stress-dependent induction of CFS genes is mediated by ubiquitin-like protein 5 (UBL5), and that a UBL5-FANCD2 dependent axis regulates the mitochondrial UPR in human cells. We propose that FANCD2 coordinates nuclear and mitochondrial activities to prevent genome instability.


Subject(s)
Chromosome Fragile Sites , Chromosome Fragility , Fanconi Anemia Complementation Group D2 Protein/genetics , Mitochondria/genetics , Stress, Physiological , DNA Damage , Fanconi Anemia Complementation Group D2 Protein/metabolism , Gene Expression Regulation , HCT116 Cells , Humans , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Phosphorylation , Transcription, Genetic , Ubiquitins/genetics , Ubiquitins/metabolism , Unfolded Protein Response
9.
Genes (Basel) ; 11(6)2020 06 10.
Article in English | MEDLINE | ID: mdl-32532049

ABSTRACT

Chromosomal instability (CIN) is associated with many human diseases, including neurodevelopmental or neurodegenerative conditions, age-related disorders and cancer, and is a key driver for disease initiation and progression. A major source of structural chromosome instability (s-CIN) leading to structural chromosome aberrations is "replication stress", a condition in which stalled or slowly progressing replication forks interfere with timely and error-free completion of the S phase. On the other hand, mitotic errors that result in chromosome mis-segregation are the cause of numerical chromosome instability (n-CIN) and aneuploidy. In this review, we will discuss recent evidence showing that these two forms of chromosomal instability can be mechanistically interlinked. We first summarize how replication stress causes structural and numerical CIN, focusing on mechanisms such as mitotic rescue of replication stress (MRRS) and centriole disengagement, which prevent or contribute to specific types of structural chromosome aberrations and segregation errors. We describe the main outcomes of segregation errors and how micronucleation and aneuploidy can be the key stimuli promoting inflammation, senescence, or chromothripsis. At the end, we discuss how CIN can reduce cellular fitness and may behave as an anticancer barrier in noncancerous cells or precancerous lesions, whereas it fuels genomic instability in the context of cancer, and how our current knowledge may be exploited for developing cancer therapies.


Subject(s)
Chromosomal Instability/genetics , Chromosome Segregation/genetics , DNA Replication/genetics , Mitosis/genetics , Aneuploidy , Centrioles/genetics , DNA Damage/genetics , Humans
10.
Redox Biol ; 36: 101618, 2020 09.
Article in English | MEDLINE | ID: mdl-32863220

ABSTRACT

Fanconi Anemia (FA) is a disease characterized by bone marrow (BM) failure and aplastic anemia. In addition to a defective DNA repair system, other mechanisms are involved in its pathogenesis, such as defective mitochondrial metabolism, accumulation of lipids, and increment of oxidative stress production. To better understand the role of these metabolic alterations in the process of HSC maturation in FA, we evaluated several biochemical and cellular parameters on mononuclear cells isolated from the bone marrow of FA patients or healthy donors. To mimic the cellular residence in the BM niche or their exit from the BM niche to the bloodstream, cells have been grown in hypoxic or normoxic conditions, respectively. The data show that, in normoxic conditions, a switch from anaerobic to aerobic metabolism occurs both in healthy and in pathological samples. However, in FA cells this change is associated with altered oxidative phosphorylation, the increment of oxidative stress production, no activation of the endogenous antioxidant defenses and arrest in the G2M phase of the cell cycle. By contrast, FA cells grown in hypoxic conditions do not show cell cycle and metabolic alterations in comparison to the healthy control, maintaining both an anaerobic flux. The data reported herein suggests that the passage from the BM niche to the bloodstream represents a crucial point in the FA pathogenesis associated with mitochondrial dysfunction.


Subject(s)
Fanconi Anemia , Bone Marrow/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Humans , Mitochondria/metabolism , Oxidative Phosphorylation , Oxidative Stress
11.
Oncotarget ; 10(43): 4407-4423, 2019 Jul 09.
Article in English | MEDLINE | ID: mdl-31320994

ABSTRACT

Dicer, an endoribonuclease best-known for its role in microRNA biogenesis and RNA interference pathway, has been shown to play a role in the DNA damage response and repair of double-stranded DNA breaks (DSBs) in mammalian cells. However, it remains unknown whether Dicer is also important to preserve genome integrity upon replication stress. To address this question, we focused our study on common fragile sites (CFSs), which are susceptible to breakage after replication stress. We show that inhibition of the Dicer pathway leads to an increase in CFS expression upon induction of replication stress and to an accumulation of 53BP1 nuclear bodies, indicating transmission of replication-associated damage. We also show that in absence of a functional Dicer or Drosha, the assembly into nuclear foci of the Fanconi anemia (FA) protein FANCD2 and of the replication and checkpoint factor TopBP1 in response to replication stress is impaired, and the activation of the S-phase checkpoint is defective. Based on these results, we propose that Dicer pre-vents genomic instability after replication stress, by allowing the proper recruitment to stalled forks of proteins that are necessary to maintain replication fork stability and activate the S-phase checkpoint, thus limiting cells from proceeding into mitosis with under-replicated DNA.

12.
Mol Biol Cell ; 15(11): 5053-63, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15371536

ABSTRACT

The mechanisms underlying completion of cytokinesis are still poorly understood. Here, we show that the Drosophila orthologue of mammalian Citron kinases is essential for the final events of the cytokinetic process. Flies bearing mutations in the Drosophila citron kinase (dck) gene were defective in both neuroblast and spermatocyte cytokinesis. In both cell types, early cytokinetic events such as central spindle assembly and contractile ring formation were completely normal. Moreover, cytokinetic rings constricted normally, leading to complete furrow ingression. However late telophases of both cell types displayed persistent midbodies associated with disorganized F actin and anillin structures. Similar defects were observed in dck RNA interference (RNAi) telophases, which, in addition to abnormal F actin and anillin rings, also displayed aberrant membrane protrusions at the cleavage site. Together, these results indicate that mutations in the dck gene result in morphologically abnormal intercellular bridges and in delayed resolution of these structures, suggesting that the wild-type function of dck is required for abscission at the end of cytokinesis. The phenotype of Dck-depleted cells is different from those observed in most Drosophila cytokinesis mutants but extraordinarily similar to that caused by anillin RNAi, suggesting that Dck and anillin are in the same pathway for completion of cytokinesis.


Subject(s)
Cytokinesis , Protein Serine-Threonine Kinases/physiology , Spermatocytes/cytology , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , Cell Membrane/metabolism , DNA/metabolism , Drosophila melanogaster , Intracellular Signaling Peptides and Proteins , Male , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Neurons/metabolism , Phenotype , Phylogeny , Protein Serine-Threonine Kinases/genetics , RNA Interference , Sequence Homology, Amino Acid , Telophase
13.
Cell Cycle ; 16(7): 613-633, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28166452

ABSTRACT

Genomic instability is a hallmark of cancer and a common feature of human disorders, characterized by growth defects, neurodegeneration, cancer predisposition, and aging. Recent evidence has shown that DNA replication stress is a major driver of genomic instability and tumorigenesis. Cells can undergo mitosis with under-replicated DNA or unresolved DNA structures, and specific pathways are dedicated to resolving these structures during mitosis, suggesting that mitotic rescue from replication stress (MRRS) is a key process influencing genome stability and cellular homeostasis. Deregulation of MRRS following oncogene activation or loss-of-function of caretaker genes may be the cause of chromosomal aberrations that promote cancer initiation and progression. In this review, we discuss the causes and consequences of replication stress, focusing on its persistence in mitosis as well as the mechanisms and factors involved in its resolution, and the potential impact of incomplete replication or aberrant MRRS on tumorigenesis, aging and disease.


Subject(s)
DNA Replication , Mitosis , Stress, Physiological , Animals , Chromosomal Instability/genetics , Chromosome Fragile Sites/genetics , Fanconi Anemia/genetics , Humans
14.
Eur J Hum Genet ; 23(7): 957-62, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25315659

ABSTRACT

SASH1 (SAM and SH3 domain-containing protein 1) is a tumor suppressor gene involved in the tumorigenesis of a spectrum of solid cancers. Heterozygous SASH1 variants are known to cause autosomal-dominant dyschromatosis. Homozygosity mapping and whole-exome sequencing were performed in a consanguineous Moroccan family with two affected siblings presenting an unclassified phenotype associating an abnormal pigmentation pattern (hypo- and hyperpigmented macules of the trunk and face and areas of reticular hypo- and hyperpigmentation of the extremities), alopecia, palmoplantar keratoderma, ungueal dystrophy and recurrent spinocellular carcinoma. We identified a homozygous variant in SASH1 (c.1849G>A; p.Glu617Lys) in both affected individuals. Wound-healing assay showed that the patient's fibroblasts were better able than control fibroblasts to migrate. Following the identification of SASH1 heterozygous variants in dyschromatosis, we used reverse phenotyping to show that autosomal-recessive variants of this gene could be responsible for an overlapping but more complex phenotype that affected skin appendages. SASH1 should be added to the list of genes responsible for autosomal-dominant and -recessive genodermatosis, with no phenotype in heterozygous patients in the recessive form, and to the list of genes responsible for a predisposition to skin cancer.


Subject(s)
Genetic Predisposition to Disease/genetics , Keratoderma, Palmoplantar/genetics , Mutation, Missense , Pigmentation Disorders/genetics , Skin Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Adult , Consanguinity , DNA Mutational Analysis/methods , Exome/genetics , Family Health , Female , Fibroblasts/metabolism , Genes, Recessive , Homozygote , Humans , Male , Pedigree , Sequence Analysis, DNA , Siblings , Skin/metabolism , Skin/pathology
15.
Nat Cell Biol ; 15(8): 1008-15, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23811686

ABSTRACT

Chromosomal instability (CIN) is a hallmark of tumour initiation and progression. Some genomic regions are particularly unstable under replication stress, notably common fragile sites (CFSs) whose rearrangements in tumour cells contribute to cancer development. Recent work has shown that the Fanconi anaemia (FANC) pathway plays a role in preventing defective chromosome segregation and CIN under conditions of replication stress. Strikingly, FANCD2 is recruited to regions hosting CFSs on metaphase chromosomes. To decipher the mechanisms protecting CFSs in G2/M, we searched for proteins that co-localize with FANCD2 on mitotic chromosomes, and identified XPF-ERCC1 and MUS81-EME1, two structure-specific endonucleases. We show that depletion of either ERCC1 or MUS81-EME1 affects accurate processing of replication intermediates or under-replicated DNA that persist at CFSs until mitosis. Depletion of these endonucleases also leads to an increase in the frequency of chromosome bridges during anaphase that, in turn, favours accumulation of DNA damage in the following G1 phase.


Subject(s)
Chromatids/metabolism , Chromosome Fragile Sites/physiology , DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Mitosis/physiology , Blotting, Western , Cell Line , Chromosomal Instability , DNA Breaks , DNA-Binding Proteins/genetics , Down-Regulation , Endonucleases/genetics , HeLa Cells , Humans , Membrane Proteins , Microscopy, Confocal , Models, Biological , Neoplasm Proteins , RNA, Small Interfering
16.
J Cell Biol ; 201(3): 395-408, 2013 Apr 29.
Article in English | MEDLINE | ID: mdl-23609533

ABSTRACT

Human DNA polymerase η (Pol η) is best known for its role in responding to UV irradiation-induced genome damage. We have recently observed that Pol η is also required for the stability of common fragile sites (CFSs), whose rearrangements are considered a driving force of oncogenesis. Here, we explored the molecular mechanisms underlying this newly identified role. We demonstrated that Pol η accumulated at CFSs upon partial replication stress and could efficiently replicate non-B DNA sequences within CFSs. Pol η deficiency led to persistence of checkpoint-blind under-replicated CFS regions in mitosis, detectable as FANCD2-associated chromosomal sites that were transmitted to daughter cells in 53BP1-shielded nuclear bodies. Expression of a catalytically inactive mutant of Pol η increased replication fork stalling and activated the replication checkpoint. These data are consistent with the requirement of Pol η-dependent DNA synthesis during S phase at replication forks stalled in CFS regions to suppress CFS instability by preventing checkpoint-blind under-replicated DNA in mitosis.


Subject(s)
Chromosome Fragile Sites , DNA-Directed DNA Polymerase/metabolism , Cell Line, Tumor , Cell Nucleus/enzymology , Chromosome Fragility , DNA Replication , DNA, B-Form/genetics , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/physiology , Homologous Recombination , Humans , Inverted Repeat Sequences , Mitosis , Protein Binding , S Phase Cell Cycle Checkpoints , Stress, Physiological
17.
Cell Cycle ; 11(23): 4385-9, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23159853

ABSTRACT

Megakaryocyte is the naturally polyploid cell that gives rise to platelets. Polyploidization occurs by endomitosis, a process corresponding to a late failure of cytokinesis with a backward movement of the daughter cells. Generally, a pure defect in cytokinesis produces a multinucleated cell, but megakaryocytes are characterized by a single polylobulated nucleus with a 2 (N) ploidy. Here, we show the existence of a defect in karyokinesis during the endomitotic process. From late telophase until the reversal of cytokinesis, some dipolar mitosis/endomitosis and most multipolar endomitosis present a thin DNA link between the segregated chromosomes surrounded by an incomplete nuclear membrane formation, which implies that sister chromatid separation is not complete. This observation may explain why polyploid megakaryocytes display a single polylobulated nucleus along with an increase in ploidy.


Subject(s)
Megakaryocytes/cytology , Mitosis , Antigens, CD34/metabolism , Cell Nucleus Division , Humans , Megakaryocytes/metabolism , Microscopy, Confocal , Telophase
18.
Nat Cell Biol ; 11(6): 761-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19465921

ABSTRACT

Loss-of-function of caretaker genes characterizes a group of cancer predisposition diseases that feature cellular hypersensitivity to DNA damage and chromosome fragility; this group includes Fanconi anaemia and Bloom syndrome. The products of the 13 FANC genes (mutated in Fanconi anaemia), which constitute the 'FANC' pathway, and BLM (the RecQ helicase mutated in Bloom syndrome) are thought to collaborate during the S phase of the cell cycle, preventing chromosome instability. Recently, BLM has been implicated in the completion of sister chromatid separation during mitosis, a complex process in which precise regulation and execution is crucial to preserve genomic stability. Here we show for the first time a role for the FANC pathway in chromosome segregation during mitotic cell division. FANCD2, a key component of the pathway, localizes to discrete spots on mitotic chromosomes. FANCD2 chromosomal localization is responsive to replicative stress and specifically targets aphidicolin (APH)-induced chromatid gaps and breaks. Our data indicate that the FANC pathway is involved in rescuing abnormal anaphase and telophase (ana-telophase) cells, limiting aneuploidy and reducing chromosome instability in daughter cells. We further address a cooperative role for the FANC pathway and BLM in preventing micronucleation, through FANC-dependent targeting of BLM to non-centromeric abnormal structures induced by replicative stress. We reveal new crosstalk between FANC and BLM proteins, extending their interaction beyond the S-phase rescue of damaged DNA to the safeguarding of chromosome stability during mitosis.


Subject(s)
Chromosome Aberrations , Fanconi Anemia Complementation Group Proteins/metabolism , Micronuclei, Chromosome-Defective , Mitosis/physiology , RecQ Helicases/metabolism , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Cell Line , Chromosome Segregation , Cytokinesis/physiology , DNA Replication , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Histones/genetics , Histones/metabolism , Humans , RNA Interference , RecQ Helicases/genetics , Signal Transduction/physiology
19.
Cell Cycle ; 8(18): 2907-11, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19729998

ABSTRACT

Fanconi anemia (FA) is a chromosome instability syndrome characterized by progressive bone marrow failure and cancer proneness. The proteins mutated in FA constitute the so-called FANC/BRCA pathway, involved in DNA replication and damage response. However, it is not completely understood how the FANC proteins perform their functions and maintain chromosome stability. Two recently published works reported that FANCD2 localizes to discrete sites on mitotic chromosomes, as consequence of replication fork stalling. The FANC pathway proved to be required to promote BLM-mediated anaphase resolution of chromosome entanglements induced by replication stress. It has also been shown that chromosome entanglement derives from DNA intertwining at fragile sites and that FANCD2 specifically targets these sites. Collectively, our data highlight a new role for the FANC proteins in the prevention of chromosome instability and aneuploidy. These findings open new directions in understanding the mechanisms of chromosome fragility and the role of FANC proteins in preserving genome stability.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/physiology , Mitosis/genetics , Chromosomal Instability , DNA Replication , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Genomic Instability
20.
Curr Biol ; 19(21): 1839-45, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19836241

ABSTRACT

In centrosome-containing cells, spindle assembly relies on microtubules (MTs) nucleated from both centrosomes and chromosomes. Recent work has suggested that additional spindle MTs can be nucleated by gamma-tubulin ring complexes (gamma-TuRCs) that associate laterally with preexisting spindle MTs, leading to spindle amplification. It has been proposed that in Drosophila S2 cells, gamma-TuRCs are anchored to the spindle MTs by augmin, a multiprotein complex that contains at least eight subunits. Here we show that the Dgt6 component of augmin is primarily required for kinetochore fiber (k-fiber) formation. An analysis of MT regrowth after cold exposure showed that formation of kinetochore-driven k-fibers is severely impaired in Dgt6-depleted cells. In control cells, these fibers are enriched in Dgt6, gamma-tubulin, and Msps/XMAP215. Consistent with these results, Dgt6 coprecipitates with Msps, D-TACC, gamma-tubulin, Ndc80, and Nuf2. However, RNA interference (RNAi)-mediated inhibition of gamma-tubulin, Msps/XMAP215, or Ndc80/Hec1 reduced but did not abolish k-fiber regrowth. These results indicate that Dgt6 plays a pivotal role in kinetochore-driven k-fiber formation, mediating nucleation and/or initial stabilization of chromosome-induced MTs. We propose that Dgt6 binds and stabilizes nascent chromatin-induced MTs, facilitating their interaction with the Ndc80-Nuf2 complex. Dgt6 may also promote elongation of kinetochore-driven k-fibers through its interaction with gamma-tubulin and Msps.


Subject(s)
Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Drosophila/metabolism , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/physiology , Microtubules/metabolism , Tubulin/metabolism , Animals , Chromosome Segregation/genetics , Chromosomes/metabolism , Drosophila/genetics , Drosophila/ultrastructure , Drosophila Proteins/analysis , Drosophila Proteins/genetics , Kinetochores/physiology , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/genetics , Spindle Apparatus/metabolism , Tubulin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL