Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Mol Cell ; 84(9): 1684-1698.e9, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38593805

ABSTRACT

The Bloom syndrome (BLM) helicase is critical for alternative lengthening of telomeres (ALT), a homology-directed repair (HDR)-mediated telomere maintenance mechanism that is prevalent in cancers of mesenchymal origin. The DNA substrates that BLM engages to direct telomere recombination during ALT remain unknown. Here, we determine that BLM helicase acts on lagging strand telomere intermediates that occur specifically in ALT-positive cells to assemble a replication-associated DNA damage response. Loss of ATRX was permissive for BLM localization to ALT telomeres in S and G2, commensurate with the appearance of telomere C-strand-specific single-stranded DNA (ssDNA). DNA2 nuclease deficiency increased 5'-flap formation in a BLM-dependent manner, while telomere C-strand, but not G-strand, nicks promoted ALT. These findings define the seminal events in the ALT DNA damage response, linking aberrant telomeric lagging strand DNA replication with a BLM-directed HDR mechanism that sustains telomere length in a subset of human cancers.


Subject(s)
DNA Damage , DNA Replication , RecQ Helicases , Telomere Homeostasis , Telomere , RecQ Helicases/metabolism , RecQ Helicases/genetics , Humans , Telomere/metabolism , Telomere/genetics , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/genetics , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , DNA Helicases/metabolism , DNA Helicases/genetics , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Bloom Syndrome/enzymology , Bloom Syndrome/pathology , Cell Line, Tumor
2.
PLoS One ; 18(6): e0281524, 2023.
Article in English | MEDLINE | ID: mdl-37267408

ABSTRACT

Bloom syndrome helicase (BLM) is a RecQ-family helicase implicated in a variety of cellular processes, including DNA replication, DNA repair, and telomere maintenance. Mutations in human BLM cause Bloom syndrome (BS), an autosomal recessive disorder that leads to myriad negative health impacts including a predisposition to cancer. BS-causing mutations in BLM often negatively impact BLM ATPase and helicase activity. While BLM mutations that cause BS have been well characterized both in vitro and in vivo, there are other less studied BLM mutations that exist in the human population that do not lead to BS. Two of these non-BS mutations, encoding BLM P868L and BLM G1120R, when homozygous, increase sister chromatid exchanges in human cells. To characterize these naturally occurring BLM mutant proteins in vitro, we purified the BLM catalytic core (BLMcore, residues 636-1298) with either the P868L or G1120R substitution. We also purified a BLMcore K869A K870A mutant protein, which alters a lysine-rich loop proximal to the P868 residue. We found that BLMcore P868L and G1120R proteins were both able to hydrolyze ATP, bind diverse DNA substrates, and unwind G-quadruplex and duplex DNA structures. Molecular dynamics simulations suggest that the P868L substitution weakens the DNA interaction with the winged-helix domain of BLM and alters the orientation of one lobe of the ATPase domain. Because BLMcore P868L and G1120R retain helicase function in vitro, it is likely that the increased genome instability is caused by specific impacts of the mutant proteins in vivo. Interestingly, we found that BLMcore K869A K870A has diminished ATPase activity, weakened binding to duplex DNA structures, and less robust helicase activity compared to wild-type BLMcore. Thus, the lysine-rich loop may have an important role in ATPase activity and specific binding and DNA unwinding functions in BLM.


Subject(s)
Bloom Syndrome , Humans , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Lysine , RecQ Helicases/genetics , RecQ Helicases/metabolism , DNA/metabolism , Mutant Proteins
3.
EMBO J ; 42(10): e111980, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36970874

ABSTRACT

Homologous recombination (HR) is a key DNA damage repair pathway that is tightly adjusted to the state of a cell. A central regulator of homologous recombination is the conserved helicase-containing Bloom syndrome complex, renowned for its crucial role in maintaining genome integrity. Here, we show that in Arabidopsis thaliana, Bloom complex activity is controlled by selective autophagy. We find that the recently identified DNA damage regulator KNO1 facilitates K63-linked ubiquitination of RMI1, a structural component of the complex, thereby triggering RMI1 autophagic degradation and resulting in increased homologous recombination. Conversely, reduced autophagic activity makes plants hypersensitive to DNA damage. KNO1 itself is also controlled at the level of proteolysis, in this case mediated by the ubiquitin-proteasome system, becoming stabilized upon DNA damage via two redundantly acting deubiquitinases, UBP12 and UBP13. These findings uncover a regulatory cascade of selective and interconnected protein degradation steps resulting in a fine-tuned HR response upon DNA damage.


Subject(s)
DNA-Binding Proteins , Homologous Recombination , Proteasome Endopeptidase Complex , Humans , Autophagy , Bloom Syndrome/metabolism , DNA-Binding Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism
4.
Int J Mol Sci ; 23(23)2022 Nov 26.
Article in English | MEDLINE | ID: mdl-36499126

ABSTRACT

Prostate cancer (PCa) is a common cancer and a major cause of cancer-related death worldwide in men, necessitating novel targets for cancer therapy. High expression of Bloom's syndrome protein (BLM) helicase is associated with the occurrence and development of PCa. Therefore, the identification and development of new BLM inhibitors may be a new direction for the treatment of PCa. Here, we identified a novel inhibitor by molecular docking and put it to systematic evaluation via various experiments, AO/854, which acted as a competitive inhibitor that blocked the BLM-DNA interaction. Cellular evaluation indicated that AO/854-suppressed tumor growth and metastasis in PC3 cells by enhancing DNA damage, phosphorylating Chk1/Chk2, and altering the p53 signaling pathway. Collectively, the study highlights the potential of BLM as a therapeutic target in PCa and reveals a distinct mechanism by which AO/854 competitively inhibits the function of BLM.


Subject(s)
Bloom Syndrome , Prostatic Neoplasms , Humans , Male , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Molecular Docking Simulation , RecQ Helicases/genetics , RecQ Helicases/metabolism , DNA/metabolism , DNA Damage , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics
5.
Sci Adv ; 8(5): eabk0221, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35119917

ABSTRACT

Bloom's syndrome is caused by inactivation of the BLM helicase, which functions with TOP3A and RMI1-2 (BTR complex) to dissolve recombination intermediates and avoid somatic crossing-over. We show here that crossover avoidance by BTR further requires the activity of cyclin-dependent kinase-1 (CDK1), Polo-like kinase-1 (PLK1), and the DDR mediator protein TOPBP1, which act in the same pathway. Mechanistically, CDK1 phosphorylates BLM and TOPBP1 and promotes the interaction of both proteins with PLK1. This is amplified by the ability of TOPBP1 to facilitate phosphorylation of BLM at sites that stimulate both BLM-PLK1 and BLM-TOPBP1 binding, creating a positive feedback loop that drives rapid BLM phosphorylation at the G2-M transition. In vitro, BLM phosphorylation by CDK/PLK1/TOPBP1 stimulates the dissolution of topologically linked DNA intermediates by BLM-TOP3A. Thus, we propose that the CDK1-TOPBP1-PLK1 axis enhances BTR-mediated dissolution of recombination intermediates late in the cell cycle to suppress crossover recombination and curtail genomic instability.


Subject(s)
Bloom Syndrome , Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Carrier Proteins/genetics , DNA-Binding Proteins/metabolism , Genomic Instability , Humans , Nuclear Proteins/metabolism , RecQ Helicases/genetics , RecQ Helicases/metabolism , Recombination, Genetic , Polo-Like Kinase 1
6.
Hum Mol Genet ; 31(13): 2185-2193, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35099000

ABSTRACT

Bloom syndrome (BS) is an autosomal recessive disease clinically characterized by primary microcephaly, growth deficiency, immunodeficiency and predisposition to cancer. It is mainly caused by biallelic loss-of-function mutations in the BLM gene, which encodes the BLM helicase, acting in DNA replication and repair processes. Here, we describe the gene expression profiles of three BS fibroblast cell lines harboring causative, biallelic truncating mutations obtained by single-cell (sc) transcriptome analysis. We compared the scRNA transcription profiles from three BS patient cell lines to two age-matched wild-type controls and observed specific deregulation of gene sets related to the molecular processes characteristically affected in BS, such as mitosis, chromosome segregation, cell cycle regulation and genomic instability. We also found specific upregulation of genes of the Fanconi anemia pathway, in particular FANCM, FANCD2 and FANCI, which encode known interaction partners of BLM. The significant deregulation of genes associated with inherited forms of primary microcephaly observed in our study might explain in part the molecular pathogenesis of microcephaly in BS, one of the main clinical characteristics in patients. Finally, our data provide first evidence of a novel link between BLM dysfunction and transcriptional changes in condensin complex I and II genes. Overall, our study provides novel insights into gene expression profiles in BS on an sc level, linking specific genes and pathways to BLM dysfunction.


Subject(s)
Bloom Syndrome , Microcephaly , Adenosine Triphosphatases , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , DNA Helicases , DNA-Binding Proteins/genetics , Humans , Multiprotein Complexes , RecQ Helicases/genetics , RecQ Helicases/metabolism
7.
Biochim Biophys Acta Mol Basis Dis ; 1867(6): 166106, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33631320

ABSTRACT

TOP3A promotes processing of double Holliday junction dissolution and also plays an important role in decatenation and segregation of human mtDNA. Recently, TOP3A mutations have been reported to cause Bloom syndrome-like disorder. However, whether the two function play equal roles in the disease pathogenesis is unclear. We retrospectively studied the disease progression of two siblings with Bloom-like syndrome caused by two novel mutations of TOP3A, p.Q788* and p.D479G. Beside the common clinical manifestations, our patients exhibited liver lipid storage with hepatomegaly. In cellular and molecular biological studies, TOP3A deficiency moderately increased sister chromatid exchanges and decreased cell proliferation compared with BLM or RMI2 deficiency. These changes were rescued by ectopic expression of either of the wildtype TOP3A or TOP3A-D479G. In contrast, reduced mitochondrial ATP generation and oxygen consumption rates observed in TOP3A defective cells were rescued by over-expression of the wildtype TOP3A, but not TOP3A-D479G. Considering the different impact of the TOP3A-D479G mutation on the genome stability and mitochondrial metabolism, we propose that the impaired mitochondrial metabolism plays an important role in the pathogenesis of TOP3A-deficient Bloom-like disease.


Subject(s)
Bloom Syndrome/pathology , DNA Topoisomerases, Type I/genetics , Mitochondria/pathology , Mutation , Bloom Syndrome/etiology , Bloom Syndrome/metabolism , Child, Preschool , Fatal Outcome , Female , Genomic Instability , Humans , Male , Mitochondria/metabolism , Pedigree , Retrospective Studies
8.
Sci Rep ; 10(1): 7490, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32367056

ABSTRACT

Werner Syndrome (WS) and Bloom Syndrome (BS) are disorders of DNA damage repair caused by biallelic disruption of the WRN or BLM DNA helicases respectively. Both are commonly associated with insulin resistant diabetes, usually accompanied by dyslipidemia and fatty liver, as seen in lipodystrophies. In keeping with this, progressive reduction of subcutaneous adipose tissue is commonly observed. To interrogate the underlying cause of adipose tissue dysfunction in these syndromes, CRISPR/Cas9 genome editing was used to generate human pluripotent stem cell (hPSC) lacking either functional WRN or BLM helicase. No deleterious effects were observed in WRN-/- or BLM-/- embryonic stem cells, however upon their differentiation into adipocyte precursors (AP), premature senescence emerged, impairing later stages of adipogenesis. The resulting adipocytes were also found to be senescent, with increased levels of senescent markers and senescence-associated secretory phenotype (SASP) components. SASP components initiate and reinforce senescence in adjacent cells, which is likely to create a positive feedback loop of cellular senescence within the adipocyte precursor compartment, as demonstrated in normal ageing. Such a scenario could progressively attenuate adipose mass and function, giving rise to "lipodystrophy-like" insulin resistance. Further assessment of pharmacological senolytic strategies are warranted to mitigate this component of Werner and Bloom syndromes.


Subject(s)
Adipocytes/metabolism , Bloom Syndrome , Cellular Senescence , Human Embryonic Stem Cells/metabolism , Models, Biological , Werner Syndrome , Adipocytes/pathology , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Bloom Syndrome/pathology , CRISPR-Cas Systems , Cell Line , Gene Deletion , Human Embryonic Stem Cells/pathology , Humans , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome/pathology
9.
Cell Rep ; 24(4): 947-961.e7, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30044990

ABSTRACT

Mutations in BLM helicase predispose Bloom syndrome (BS) patients to a wide spectrum of cancers. We demonstrate that MIB1-ubiquitylated BLM in G1 phase functions as an adaptor protein by enhancing the binding of transcription factor c-Jun and its E3 ligase, Fbw7α. BLM enhances the K48/K63-linked ubiquitylation on c-Jun, thereby enhancing the rate of its subsequent degradation. Functionally defective Fbw7α mutants prevalent in multiple human cancers are reactivated by BLM. However, BS patient-derived BLM mutants cannot potentiate Fbw7α-dependent c-Jun degradation. The decrease in the levels of c-Jun in cells expressing BLM prevents effective c-Jun binding to 2,584 gene promoters. This causes decreases in the transcript and protein levels of c-Jun targets in BLM-expressing cells, resulting in attenuated c-Jun-dependent effects during neoplastic transformation. Thus, BLM carries out its function as a tumor suppressor by enhancing c-Jun turnover and thereby preventing its activity as a proto-oncogene.


Subject(s)
F-Box-WD Repeat-Containing Protein 7/metabolism , Genes, jun , Proto-Oncogene Proteins c-jun/metabolism , RecQ Helicases/metabolism , Animals , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Carcinogenesis , F-Box-WD Repeat-Containing Protein 7/genetics , G1 Phase , HCT116 Cells , HEK293 Cells , Humans , Mice , Mice, Nude , Mutation , Proto-Oncogene Mas , Proto-Oncogene Proteins c-jun/genetics , RecQ Helicases/genetics , Ubiquitination
10.
J Cell Biol ; 216(12): 3991-4005, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29042409

ABSTRACT

Sgs1, the orthologue of human Bloom's syndrome helicase BLM, is a yeast DNA helicase functioning in DNA replication and repair. We show that SGS1 loss increases R-loop accumulation and sensitizes cells to transcription-replication collisions. Yeast lacking SGS1 accumulate R-loops and γ-H2A at sites of Sgs1 binding, replication pausing regions, and long genes. The mutation signature of sgs1Δ reveals copy number changes flanked by repetitive regions with high R-loop-forming potential. Analysis of BLM in Bloom's syndrome fibroblasts or by depletion of BLM from human cancer cells confirms a role for Sgs1/BLM in suppressing R-loop-associated genome instability across species. In support of a potential direct effect, BLM is found physically proximal to DNA:RNA hybrids in human cells, and can efficiently unwind R-loops in vitro. Together, our data describe a conserved role for Sgs1/BLM in R-loop suppression and support an increasingly broad view of DNA repair and replication fork stabilizing proteins as modulators of R-loop-mediated genome instability.


Subject(s)
Bloom Syndrome/genetics , DNA/chemistry , Genomic Instability , RecQ Helicases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Bloom Syndrome/metabolism , Bloom Syndrome/pathology , Cell Line, Transformed , Cell Line, Tumor , DNA/genetics , DNA/metabolism , DNA Repair , DNA Replication , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Dosage , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Humans , Nucleic Acid Conformation , Protein Binding , RNA/genetics , RNA/metabolism , RecQ Helicases/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/metabolism
11.
Annu Rev Biochem ; 86: 461-484, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28654322

ABSTRACT

Self-catalyzed DNA depurination is a sequence-specific physiological mechanism mediated by spontaneous extrusion of a stem-loop catalytic intermediate. Hydrolysis of the 5'G residue of the 5'GA/TGG loop and of the first 5'A residue of the 5'GAGA loop, together with particular first stem base pairs, specifies their hydrolysis without involving protein, cofactor, or cation. As such, this mechanism is the only known DNA catalytic activity exploited by nature. The consensus sequences for self-depurination of such G- and A-loop residues occur in all genomes examined across the phyla, averaging one site every 2,000-4,000 base pairs. Because apurinic sites are subject to error-prone repair, leading to substitution and short frameshift mutations, they are both a source of genome damage and a means for creating sequence diversity. Their marked overrepresentation in genomes, and largely unchanging density from the lowest to the highest organisms, indicate their selection over the course of evolution. The mutagenicity at such sites in many human genes is associated with loss of function of key proteins responsible for diverse diseases.


Subject(s)
Adenine/metabolism , Bloom Syndrome/genetics , DNA, Catalytic/genetics , Guanine/metabolism , Polymorphism, Genetic , Werner Syndrome/genetics , Biological Evolution , Bloom Syndrome/metabolism , Bloom Syndrome/pathology , Catalysis , DNA Repair , DNA, Catalytic/metabolism , DNA, Cruciform/genetics , DNA, Cruciform/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Humans , Hydrolysis , Inverted Repeat Sequences , Mutation , Werner Syndrome/metabolism , Werner Syndrome/pathology , beta-Globins/genetics , beta-Globins/metabolism
12.
Redox Biol ; 11: 375-383, 2017 04.
Article in English | MEDLINE | ID: mdl-28063379

ABSTRACT

Rare pleiotropic genetic disorders, Ataxia-telangiectasia (A-T), Bloom syndrome (BS) and Nijmegen breakage syndrome (NBS) are characterised by immunodeficiency, extreme radiosensitivity, higher cancer susceptibility, premature aging, neurodegeneration and insulin resistance. Some of these functional abnormalities can be explained by aberrant DNA damage response and chromosomal instability. It has been suggested that one possible common denominator of these conditions could be chronic oxidative stress caused by endogenous ROS overproduction and impairment of mitochondrial homeostasis. Recent studies indicate new, alternative sources of oxidative stress in A-T, BS and NBS cells, including NADPH oxidase 4 (NOX4), oxidised low-density lipoprotein (ox-LDL) or Poly (ADP-ribose) polymerases (PARP). Mitochondrial abnormalities such as changes in the ultrastructure and function of mitochondria, excess mROS production as well as mitochondrial damage have also been reported in A-T, BS and NBS cells. A-T, BS and NBS cells are inextricably linked to high levels of reactive oxygen species (ROS), and thereby, chronic oxidative stress may be a major phenotypic hallmark in these diseases. Due to the presence of mitochondrial disturbances, A-T, BS and NBS may be considered mitochondrial diseases. Excess activity of antioxidant enzymes and an insufficient amount of low molecular weight antioxidants indicate new pharmacological strategies for patients suffering from the aforementioned diseases. However, at the current stage of research we are unable to ascertain if antioxidants and free radical scavengers can improve the condition or prolong the survival time of A-T, BS and NBS patients. Therefore, it is necessary to conduct experimental studies in a human model.


Subject(s)
Ataxia Telangiectasia/genetics , Bloom Syndrome/genetics , DNA Repair , Mitochondria/metabolism , Nijmegen Breakage Syndrome/genetics , Oxidative Stress/genetics , Ataxia Telangiectasia/metabolism , Ataxia Telangiectasia/pathology , Bloom Syndrome/metabolism , Bloom Syndrome/pathology , DNA Damage , Gene Expression Regulation , Humans , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Mitochondria/pathology , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Nijmegen Breakage Syndrome/metabolism , Nijmegen Breakage Syndrome/pathology , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
13.
J Gastroenterol ; 52(3): 327-340, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27169843

ABSTRACT

BACKGROUND: Bloom syndrome is a rare and recessive disorder characterized by loss-of-function mutations of the BLM gene, which encodes a RecQ 3'-5' DNA helicase. Despite its putative tumor suppressor function, the contribution of BLM to human sporadic colorectal cancer (CRC) remains poorly understood. METHODS: The transcriptional regulation mechanism underlying BLM and related DNA damage response regulation in independent CRC subsets and a panel of derived cell lines was investigated by bioinformatics analysis, the transcriptomic profile, a CpG island promoter methylation assay, Western blot, and an immunolocalization assay. RESULTS: In silico analysis of gene expression data sets revealed that BLM is overexpressed in poorly differentiated CRC and exhibits a close connection with shorter relapse-free survival even after adjustment for prognostic factors and pathways that respond to DNA damage response through ataxia telangiectasia mutated (ATM) signaling. Functional characterization demonstrated that CpG island promoter hypomethylation increases BLM expression and associates with cytoplasmic BLM mislocalization and increased DNA damage response both in clinical CRC samples and in derived cancer cell lines. The DNA-damaging agent S-adenosylmethionine suppresses BLM expression, leading to the inhibition of cell growth following accumulation of DNA damage. In tumor specimens, cytoplasmic accumulation of BLM correlates with DNA damage and γH2AX and phosphorylated ATM foci and predicts long-term progression-free survival in metastatic patients treated with irinotecan. CONCLUSIONS: Taken together, the findings of this study provide the first evidence that cancer-linked DNA hypomethylation and cytosolic BLM mislocalization might reflect compromised levels of DNA-repair activity and enhanced hypersensitivity to DNA-damaging agents in CRC patients.


Subject(s)
Colorectal Neoplasms/genetics , DNA Damage , RecQ Helicases/genetics , Ataxia Telangiectasia/genetics , Bloom Syndrome/complications , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Cell Line, Tumor , Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , CpG Islands/genetics , Cytoplasm/metabolism , DNA Methylation , DNA Repair , DNA, Neoplasm/genetics , Female , Gene Expression Profiling/methods , Gene Silencing , Humans , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Staging , RNA, Messenger/genetics , RecQ Helicases/metabolism , Signal Transduction/genetics , Up-Regulation
14.
Nucleic Acids Res ; 44(14): 6787-93, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27185886

ABSTRACT

Sister chromatid exchanges (SCEs) are considered sensitive indicators of genome instability. Detection of SCEs typically requires cells to incorporate bromodeoxyuridine (BrdU) during two rounds of DNA synthesis. Previous studies have suggested that SCEs are induced by DNA replication over BrdU-substituted DNA and that BrdU incorporation alone could be responsible for the high number of SCE events observed in cells from patients with Bloom syndrome (BS), a rare genetic disorder characterized by marked genome instability and high SCE frequency. Here we show using Strand-seq, a single cell DNA template strand sequencing technique, that the presence of variable BrdU concentrations in the cell culture medium and in DNA template strands has no effect on SCE frequency in either normal or BS cells. We conclude that BrdU does not induce SCEs and that SCEs detected in either normal or BS cells reflect DNA repair events that occur spontaneously.


Subject(s)
Bloom Syndrome/metabolism , Bloom Syndrome/pathology , Bromodeoxyuridine/pharmacology , Sister Chromatid Exchange/drug effects , Cell Division/drug effects , DNA/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lymphocytes/drug effects , Lymphocytes/metabolism , Lymphocytes/pathology , Templates, Genetic
15.
Int J Mol Sci ; 16(11): 26395-405, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26556339

ABSTRACT

Chromosome instability usually leads to tumorigenesis. Bloom syndrome (BS) is a genetic disease associated with chromosome instability. The BS gene product, BLM, has been reported to function in the spindle assembly checkpoint (SAC) to prevent chromosome instability. BTR complex, composed of BLM, topoisomerase IIIα (Topo IIIα), RMI1 (RecQ-mediated genome instability protein 1, BLAP75) and RMI2 (RecQ-mediated genome instability protein 2, BLAP18), is crucial for maintaining genome stability. Recent work has demonstrated that RMI2 also plays critical role in SAC. However, little is know about RMI1 regulation during the cell cycle. Here we present that RMI1 protein level does not change through G1, S and G2 phases, but significantly increases in M phase. Moreover, phosphorylation of RMI1 occurs in mitosis. Upon microtubule-disturbing agent, RMI1 is phosphorylated primarily at the sites of Serine 284 and Serine 292, which does not interfere with the formation of BTR complex. Additionally, this phosphorylation is partially reversed by roscovitine treatment, implying cycling-dependent kinase 1 (CDK1) might be one of the upstream kinases.


Subject(s)
Carrier Proteins/metabolism , Mitosis , Nuclear Proteins/metabolism , Serine/metabolism , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , CDC2 Protein Kinase , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Cycle Proteins/metabolism , Codon , Cyclin-Dependent Kinases/metabolism , DNA Topoisomerases, Type I/metabolism , DNA-Binding Proteins , G1 Phase , G2 Phase , HeLa Cells , Humans , Multiprotein Complexes/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , RecQ Helicases/metabolism
16.
J Eur Acad Dermatol Venereol ; 28(10): 1318-23, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24118499

ABSTRACT

The Bloom syndrome (BS) is an autosomal recessive disorder associated with dwarfism, immunodeficiency, reduced fertility and cancer risk. BS cells show genomic instability, particularly an hyper exchange between the sister chromatids due to a defective processing of the DNA replication intermediates. It is caused by mutations in the BLM gene which encodes a member of the RecQ family of DExH box DNA helicases. In this study, we reported cytogenetic, BLM linkage and mutational analyses for two affected Tunisian families. The Cytogenetic parameters were performed by chromosomal aberration (CA) and sister chromatid exchange (SCE) assays and results showed a significant increase in mean frequency of CA and SCE in BS cells. BLM linkage performed by microsatellite genotyping revealed homozygous haplotypes for the BS patients, evidence of linkage to BLM gene. Mutational analysis by direct DNA sequencing revealed a novel frameshift mutation (c.1980-1982delAA) in exon 8 of BLM gene, resulting in a truncated protein (p.Lys662fsX5). The truncated protein could explain genomic instability and its related symptoms in the BS patients. The screening of this mutation is useful for BS diagnosis confirmation in Tunisian families.


Subject(s)
Bloom Syndrome/genetics , Chromosomal Instability/genetics , DNA Helicases/genetics , Frameshift Mutation , RecQ Helicases/genetics , Adolescent , Adult , Bloom Syndrome/metabolism , DNA Mutational Analysis , Female , Genotype , Humans , Male , Pedigree , RecQ Helicases/metabolism , Tunisia , Young Adult
17.
Cytogenet Genome Res ; 144(4): 255-263, 2014.
Article in English | MEDLINE | ID: mdl-25766002

ABSTRACT

Biallelic mutations in BLM cause Bloom syndrome (BS), a genome instability disorder characterized by growth retardation, sun sensitivity and a predisposition to cancer. As evidence of decreased genome stability, BS cells demonstrate not only elevated levels of spontaneous sister chromatid exchanges (SCEs), but also exhibit chromosomal radial formation. The molecular nature and mechanism of radial formation is not known, but radials have been thought to be DNA recombination intermediates between homologs that failed to resolve. However, we find that radials in BS cells occur over 95% between non-homologous chromosomes, and occur non-randomly throughout the genome. BLM must be phosphorylated at T99 and T122 for certain cell cycle checkpoints, but it is not known whether these modifications are necessary to suppress radial formation. We find that exogenous BLM constructs preventing phosphorylation at T99 and T122 are not able to suppress radial formation in BS cells, but are able to inhibit SCE formation. These findings indicate that BLM functions in 2 distinct pathways requiring different modifications. In one pathway, for which the phosphorylation marks appear dispensable, BLM functions to suppress SCE formation. In a second pathway, T99 and T122 phosphorylations are essential for suppression of chromosomal radial formation, both those formed spontaneously and those formed following interstrand crosslink damage.


Subject(s)
Bloom Syndrome/genetics , Chromosomal Instability , RecQ Helicases/metabolism , Sister Chromatid Exchange , Bloom Syndrome/metabolism , Cells, Cultured , Chromosomes, Human/genetics , Humans , Monte Carlo Method , Mutation , Phosphorylation , RecQ Helicases/genetics
18.
Proc Natl Acad Sci U S A ; 110(26): 10646-51, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23509288

ABSTRACT

The Bloom syndrome gene product, BLM, is a member of the highly conserved RecQ family. An emerging concept is the BLM helicase collaborates with the homologous recombination (HR) machinery to help avoid undesirable HR events and to achieve a high degree of fidelity during the HR reaction. However, exactly how such coordination occurs in vivo is poorly understood. Here, we identified a protein termed SPIDR (scaffolding protein involved in DNA repair) as the link between BLM and the HR machinery. SPIDR independently interacts with BLM and RAD51 and promotes the formation of a BLM/RAD51-containing complex of biological importance. Consistent with its role as a scaffolding protein for the assembly of BLM and RAD51 foci, cells depleted of SPIDR show increased rate of sister chromatid exchange and defects in HR. Moreover, SPIDR depletion leads to genome instability and causes hypersensitivity to DNA damaging agents. We propose that, through providing a scaffold for the cooperation of BLM and RAD51 in a multifunctional DNA-processing complex, SPIDR not only regulates the efficiency of HR, but also dictates the specific HR pathway.


Subject(s)
Proteins/metabolism , Rad51 Recombinase/metabolism , RecQ Helicases/metabolism , Recombinational DNA Repair/physiology , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Cell Line , DNA-Binding Proteins , HEK293 Cells , HeLa Cells , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nuclear Proteins , Protein Interaction Maps , Proteins/chemistry , Proteins/genetics , Rad51 Recombinase/chemistry , Rad51 Recombinase/genetics , RecQ Helicases/chemistry , RecQ Helicases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinational DNA Repair/genetics
19.
Mutat Res ; 743-744: 89-96, 2013.
Article in English | MEDLINE | ID: mdl-23261817

ABSTRACT

Bloom's syndrome (BS) is an inherited disorder caused by loss of function of the recQ-like BLM helicase. It is characterized clinically by severe growth retardation and cancer predisposition. BLM localizes to PML nuclear bodies and to the nucleolus; its deficiency results in increased intra- and inter-chromosomal recombination, including hyper-recombination of rDNA repeats. Our previous work has shown that BLM facilitates RNA polymerase I-mediated rRNA transcription in the nucleolus (Grierson et al., 2012 [18]). This study uses protein co-immunoprecipitation and in vitro transcription/translation (IVTT) to identify a direct interaction of DNA topoisomerase I with the C-terminus of BLM in the nucleolus. In vitro helicase assays demonstrate that DNA topoisomerase I stimulates BLM helicase activity on a nucleolar-relevant RNA:DNA hybrid, but has an insignificant effect on BLM helicase activity on a control DNA:DNA duplex substrate. Reciprocally, BLM enhances the DNA relaxation activity of DNA topoisomerase I on supercoiled DNA substrates. Our study suggests that BLM and DNA topoisomerase I function coordinately to modulate RNA:DNA hybrid formation as well as relaxation of DNA supercoils in the context of nucleolar transcription.


Subject(s)
DNA Topoisomerases, Type I/genetics , DNA Topoisomerases, Type I/metabolism , DNA, Ribosomal/genetics , RecQ Helicases/genetics , RecQ Helicases/metabolism , Transcription, Genetic , Bloom Syndrome/enzymology , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Cell Line , Cell Line, Tumor , Cell Nucleolus/enzymology , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , DNA, Ribosomal/metabolism , HEK293 Cells , Humans , MCF-7 Cells
20.
BMC Mol Biol ; 13: 33, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23110454

ABSTRACT

Defects in the human BLM gene cause Bloom syndrome, notable for early development of tumors in a broad variety of tissues. On the basis of sequence similarity, BLM has been identified as one of the five human homologs of RecQ from Escherichia coli. Nevertheless, biochemical characterization of the BLM protein indicates far greater functional similarity to the E. coli RecG protein and there is no known RecG homolog in human cells. To explore the possibility that the shared biochemistries of BLM and RecG may represent an example of convergent evolution of cellular function where in humans BLM has evolved to fulfill the genomic stabilization role of RecG, we determined whether expression of RecG in human BLM-deficient cells could suppress established functional cellular Bloom syndrome phenotypes. We found that RecG can indeed largely suppress both the definitive elevated sister chromatid exchange phenotype and the more recently demonstrated gene cluster instability phenotype of BLM-deficient cells. In contrast, expression of RecG has no impact on either of these phenotypes in human cells with functional BLM protein. These results suggest that the combination of biochemical activities shared by RecG and BLM fill the same evolutionary niche in preserving genomic integrity without requiring exactly identical molecular mechanisms.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , RecQ Helicases/metabolism , Bloom Syndrome/metabolism , Bloom Syndrome/pathology , Cell Line , Escherichia coli Proteins/genetics , HeLa Cells , Humans , Multigene Family , Phenotype , RecQ Helicases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...