Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Cell ; 37(6): 865-78, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20347428

ABSTRACT

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


Subject(s)
DNA Helicases/metabolism , DNA/metabolism , Genomic Instability , Histones/metabolism , Protein Folding , Protein Multimerization , Amino Acid Sequence , Animals , Cell Line , Chickens , DNA/genetics , DNA Damage , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Evolution, Molecular , Fanconi Anemia Complementation Group Proteins , Humans , Molecular Sequence Data , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Sequence Alignment , Sister Chromatid Exchange
2.
Mol Cell Biol ; 30(4): 935-47, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19995907

ABSTRACT

Tight regulation of microtubule (MT) dynamics is essential for proper chromosome movement during mitosis. Here we show, using mammalian cells, that structure-specific recognition protein 1 (SSRP1) is a novel regulator of MT dynamics. SSRP1 colocalizes with the spindle and midbody MTs, and associates with MTs both in vitro and in vivo. Purified SSRP1 facilitates tubulin polymerization and MT bundling in vitro. Knockdown of SSRP1 inhibits the growth of MTs and leads to disorganized spindle structures, reduction of K-fibers and midbody fibers, disrupted chromosome movement, and attenuated cytokinesis in vivo. These results demonstrate that SSRP1 is crucial for MT growth and spindle assembly during mitosis.


Subject(s)
DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Microtubules/metabolism , Mitosis , Transcriptional Elongation Factors/metabolism , Cell Line , Chromosome Segregation , DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Humans , Microscopy, Electron, Transmission , Microtubules/ultrastructure , Protein Binding , Spindle Apparatus/metabolism , Transcriptional Elongation Factors/genetics
3.
J Biol Chem ; 284(38): 25560-8, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19633289

ABSTRACT

Genomic stability requires a functional Fanconi anemia (FA) pathway composed of an upstream "core complex" (FA proteins A/B/C/E/F/G/L/M) that mediates monoubiquitination of the downstream targets FANCD2 and FANCI. Unique among FA core complex members, FANCM has processing activities toward replication-associated DNA structures, suggesting a vital role for FANCM during replication. Using Xenopus egg extracts, we analyzed the functions of FANCM in replication and the DNA damage response. xFANCM binds chromatin in a replication-dependent manner and is phosphorylated in response to DNA damage structures. Chromatin binding and DNA damage-induced phosphorylation of xFANCM are mediated in part by the downstream FA pathway protein FANCD2. Moreover, phosphorylation and chromatin recruitment of FANCM is regulated by two mayor players in the DNA damage response: the cell cycle checkpoint kinases ATR and ATM. Our results indicate that functions of FANCM are controlled by FA- and non-FA pathways in the DNA damage response.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage/physiology , DNA Helicases/metabolism , DNA Replication/physiology , DNA-Binding Proteins/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Oocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Xenopus Proteins/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Chromatin , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Oocytes/cytology , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis
4.
Int J Cancer ; 124(4): 783-92, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19048618

ABSTRACT

The Fanconi Anemia (FA) DNA damage response pathway is involved in the processing of DNA interstrand crosslinks (ICLs). As such, inhibition of the FA pathway could chemosensitize FA-competent tumor cells to commonly used ICL agents like cisplatin. Moreover, suppression of the FA pathway is synthetic lethal with deficiencies in several other DNA repair pathways, suggesting that FA pathway inhibitors could be used in targeted therapies against specific tumors. To identify such inhibitors, we designed a novel in vitro screening assay utilizing Xenopus egg extracts. Using the DNA-stimulated monoubiquitylation of Xenopus FANCD2 (xFANCD2-L) as readout, a chemical library screen identified DDN (2,3-dichloro-5,8-dihydroxy-1,4-naphthoquinone) as a novel and potent FA pathway inhibitor. DDN inhibited xFANCD2-L formation in a dose-dependent manner in both extracts and human cells without disruption of the upstream FA core complex. DDN also inhibited the characteristic subnuclear FANCD2 foci formation following DNA damage. Moreover, DDN displayed a greater synergistic effect with cisplatin in a FA-proficient cancer cell line compared to its FA-deficient isogenic counterpart, suggesting that DDN might be a good lead candidate as cisplatin chemosensitizer in both FA-deficient and FA-competent tumors. This system constitutes the first cell-free screening assay for identifying compounds that inhibit the FA pathway and provides a new biochemical platform for mapping the functions of its various components with specific chemical inhibitors.


Subject(s)
Drug Evaluation, Preclinical/instrumentation , Drug Evaluation, Preclinical/methods , Fanconi Anemia/drug therapy , Fanconi Anemia/genetics , Animals , Cell Survival , Cell-Free System , Cisplatin/pharmacology , Cross-Linking Reagents/pharmacology , DNA/chemistry , DNA Damage , DNA Repair , Fanconi Anemia Complementation Group D2 Protein/metabolism , HeLa Cells , Humans , Models, Biological , Xenopus laevis
5.
DNA Repair (Amst) ; 7(12): 1973-81, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18786657

ABSTRACT

Fanconi anemia (FA) is a recessive genetic disorder characterized by hypersensitivity to crosslinking agents that has been attributed to defects in DNA repair and/or replication. FANCD2 and the FA core complex bind to chromatin during DNA replication; however, the role of FA proteins during replication is unknown. Using Xenopus cell-free extracts, we show that FANCL depletion results in defective DNA replication restart following treatment with camptothecin, a drug that results in DSBs during DNA replication. This defect is more pronounced following treatment with mitomycin C, presumably because of an additional role of the FA pathway in DNA crosslink repair. Moreover, we show that chromatin binding of FA core complex proteins during DNA replication follows origin assembly and origin firing and is dependent on the binding of RPA to ssDNA while FANCD2 additionally requires ATR, consistent with FA proteins acting at replication forks. Together, our data suggest that FA proteins play a role in replication restart at collapsed replication forks.


Subject(s)
Chromatin/metabolism , DNA Replication , Fanconi Anemia Complementation Group D2 Protein/physiology , Fanconi Anemia Complementation Group L Protein/physiology , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Aphidicolin/pharmacology , Ataxia Telangiectasia Mutated Proteins , Camptothecin/pharmacology , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Cell-Free System , DNA Damage , DNA Repair , Enzyme Inhibitors/pharmacology , Fanconi Anemia , Mitomycin/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Xenopus Proteins/metabolism , Xenopus laevis
6.
Genes Cells ; 12(7): 841-51, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17584296

ABSTRACT

Fanconi anemia (FA) is associated with variable developmental abnormalities, bone marrow failure and cancer susceptibility. FANCG/XRCC9 is member of the FA core complex, a group of proteins that control the monoubiquitylation of FANCD2, an event that plays a critical role in maintaining genomic stability. Here we report the identification of the Xenopus laevis ortholog of human FANCG (xFANCG), its expression during development, and its molecular interactions with a partner protein, xFANCA. The xFANCG protein sequence is 47% similar to its human ortholog, with highest conservation in the two putative N-terminal leucine zippers and the tetratricopeptide repeat (TPR) motifs. xFANCG is maternally and zygotically transcribed. Prior to the midblastula stage, a single xFANCG transcript is observed but two additional alternatively spliced mRNAs are detected after the midblastula transition. One of the variants is predicted to encode a novel isoform of xFANCG lacking exon 2. The mutual association between FANCG and FANCA required for their nuclear import is conserved in Xenopus egg extracts. Our data demonstrate that interactions between FANCA and FANCG occur at the earliest stage of vertebrate development and raise the possibility that functionally different isoforms of xFANCG may play a role in early development.


Subject(s)
Fanconi Anemia Complementation Group G Protein/genetics , Gene Expression Regulation, Developmental , Xenopus laevis/genetics , Amino Acid Sequence , Animals , Base Sequence , Conserved Sequence , DNA, Complementary/isolation & purification , Embryo, Nonmammalian , Exons , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group G Protein/isolation & purification , Fanconi Anemia Complementation Group G Protein/metabolism , Humans , Molecular Sequence Data , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/isolation & purification , RNA, Messenger, Stored/isolation & purification , RNA, Messenger, Stored/metabolism , Sequence Homology , Xenopus laevis/embryology
7.
Mol Cell Biol ; 26(2): 425-37, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16382135

ABSTRACT

Fanconi anemia (FA) is a multigene cancer susceptibility disorder characterized by cellular hypersensitivity to DNA interstrand cross-linking agents such as mitomycin C (MMC). FA proteins are suspected to function at the interface between cell cycle checkpoints, DNA repair, and DNA replication. Using replicating extracts from Xenopus eggs, we developed cell-free assays for FA proteins (xFA). Recruitment of the xFA core complex and xFANCD2 to chromatin is strictly dependent on replication initiation, even in the presence of MMC indicating specific recruitment to DNA lesions encountered by the replication machinery. The increase in xFA chromatin binding following treatment with MMC is part of a caffeine-sensitive S-phase checkpoint that is controlled by xATR. Recruitment of xFANCD2, but not xFANCA, is dependent on the xATR-xATR-interacting protein (xATRIP) complex. Immunodepletion of either xFANCA or xFANCD2 from egg extracts results in accumulation of chromosomal DNA breaks during replicative synthesis. Our results suggest coordinated chromatin recruitment of xFA proteins in response to replication-associated DNA lesions and indicate that xFA proteins function to prevent the accumulation of DNA breaks that arise during unperturbed replication.


Subject(s)
Carrier Proteins/metabolism , DNA Damage/physiology , DNA Replication , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group D2 Protein/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Xenopus Proteins/metabolism , Amino Acid Sequence , Animals , Ataxia Telangiectasia Mutated Proteins , Caffeine/pharmacology , Cell Cycle Proteins/metabolism , Chromatin/metabolism , Cross-Linking Reagents/pharmacology , DNA Repair/physiology , Female , In Vitro Techniques , Mitomycin/pharmacology , Molecular Sequence Data , Oocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , S Phase/drug effects , S Phase/physiology , Sequence Homology, Amino Acid , Xenopus laevis
8.
Nat Genet ; 37(9): 958-63, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16116422

ABSTRACT

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


Subject(s)
Archaea/chemistry , DNA Helicases/genetics , DNA Repair , Fanconi Anemia/genetics , Hemagglutinins, Viral/genetics , Ligases/genetics , Viral Fusion Proteins/genetics , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Biological Evolution , DNA/metabolism , DNA Helicases/deficiency , DNA Helicases/metabolism , Fanconi Anemia/enzymology , Fanconi Anemia Complementation Group D2 Protein , Fanconi Anemia Complementation Group L Protein , Humans , Immunoprecipitation , Ligases/deficiency , Ligases/metabolism , Molecular Sequence Data , Mutation , Nuclear Proteins/metabolism , Phosphorylation , Protein Transport , Ubiquitin/metabolism , Viral Fusion Proteins/deficiency
9.
J Biol Chem ; 277(29): 26327-34, 2002 Jul 19.
Article in English | MEDLINE | ID: mdl-11986317

ABSTRACT

FAZF, a member of the BTB/POZ family of transcriptional repressor proteins, has been shown to bind to FANCC, the protein defective in patients with the bone marrow failure syndrome Fanconi anemia complementation group C. Because bone marrow failure in Fanconi anemia has been attributed to a failure of the hematopoietic stem cell population to produce sufficient progeny, we documented the expression of FAZF in human CD34(+) hematopoietic progenitor cells. FAZF was expressed at high levels in early stages of differentiation but declined during subsequent differentiation into erythroid and myeloid lineages. Consistent with its presumed role as a transcriptional repressor, FAZF was found in the nuclear compartment, where it resides in distinct nuclear speckles at or near sites of DNA replication. Using a FAZF-inducible myeloid cell line, we found that enforced expression of FAZF was accompanied by accumulation in the G(1) phase of the cell cycle followed later by apoptosis. These results suggest an essential role for FAZF during the proliferative stages of primitive hematopoietic progenitors, possibly acting in concert with (a subset of) the Fanconi anemia proteins.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , DNA-Binding Proteins/physiology , Repressor Proteins , Zinc Fingers/physiology , Antigens, CD34/analysis , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , DNA Replication , G1 Phase , Hematopoietic Stem Cells/metabolism , Humans , Oligopeptides , Peptides , Reverse Transcriptase Polymerase Chain Reaction
10.
Genes Cells ; 7(3): 333-42, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11918676

ABSTRACT

BACKGROUND: Fanconi anaemia (FA) is an autosomal recessive chromosomal instability disorder. Six distinct FA disease genes have been identified, the products of which function in an integrated pathway that is thought to support a nuclear caretaker function. Comparison of FA gene characteristics in different species may help to unravel the molecular function of the FA pathway. RESULTS: We have cloned the murine homologue of the Fanconi anaemia complementation group G gene, FANCG/XRCC9. The murine Fancg protein shows an 83% similarity to the human protein sequence, and has a predicted molecular weight of 68.5 kDa. Expression of mouse Fancg in human FA-G lymphoblasts fully corrects their cross-linker hypersensitivity. At mRNA and protein levels we detected the co-expression of Fancg and Fanca in murine tissues. In addition, mouse Fancg and Fanca proteins co-purify by immunoprecipitation. Upon transfection into Fanca-deficient mouse embryonic fibroblasts EGFP-Fancg chimeric protein was detectable in the nucleus. CONCLUSIONS: We identified a murine cDNA, Fancg, which cross-complements the cellular defect of human FA-G cells and thus represents a true homologue of human FANCG. Spleen, thymus and testis showed the highest Fancg expression levels. Although Fancg and Fanca are able to form a complex, this interaction is not required for Fancg to accumulate in the nuclear compartment.


Subject(s)
DNA-Binding Proteins/metabolism , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Amino Acid Sequence , Animals , DNA-Binding Proteins/genetics , Fanconi Anemia Complementation Group A Protein , Fanconi Anemia Complementation Group G Protein , Fibroblasts/metabolism , Green Fluorescent Proteins , Luminescent Proteins , Mice , Molecular Sequence Data , Proteins/metabolism , RNA, Messenger , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...