Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
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.
Mol Med Rep ; 27(5)2023 May.
Article in English | MEDLINE | ID: mdl-37052241

ABSTRACT

Pathogenic variants affecting the BLM gene are responsible for the manifestation of extremely rare cancer­predisposing Bloom syndrome. The present study reports on a case of an infant with a congenital hypotrophy, short stature and abnormal facial appearance. Initially she was examined using a routine molecular diagnostic algorithm, including the cytogenetic analysis of her karyotype, microarray analysis and methylation­specific MLPA, however, she remained undiagnosed on a molecular level. Therefore, she and her parents were enrolled in the project of trio­based exome sequencing (ES) using Human Core Exome kit. She was revealed as a carrier of an extremely rare combination of causative sequence variants altering the BLM gene (NM_000057.4), c.1642C>T and c.2207_2212delinsTAGATTC in the compound heterozygosity, resulting in a diagnosis of Bloom syndrome. Simultaneously, a mosaic loss of heterozygosity of chromosome 11p was detected and then confirmed as a borderline imprinting center 1 hypermethylation on chromosome 11p15. The diagnosis of Bloom syndrome and mosaic copy­number neutral loss of heterozygosity of chromosome 11p increases a lifetime risk to develop any types of malignancy. This case demonstrates the trio­based ES as a complex approach for the molecular diagnostics of rare pediatric diseases.


Subject(s)
Bloom Syndrome , Humans , Child , Infant , Female , Male , Bloom Syndrome/diagnosis , Bloom Syndrome/genetics , Bloom Syndrome/pathology , Exome Sequencing , Chromosomes, Human, Y , Mosaicism , Heterozygote
5.
Nat Commun ; 12(1): 4255, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34253720

ABSTRACT

Homology-directed repair (HDR), a critical DNA repair pathway in mammalian cells, is complex, leading to multiple outcomes with different impacts on genomic integrity. However, the factors that control these different outcomes are often not well understood. Here we show that SWS1-SWSAP1-SPIDR controls distinct types of HDR. Despite their requirement for stable assembly of RAD51 recombinase at DNA damage sites, these proteins are not essential for intra-chromosomal HDR, providing insight into why patients and mice with mutations are viable. However, SWS1-SWSAP1-SPIDR is critical for inter-homolog HDR, the first mitotic factor identified specifically for this function. Furthermore, SWS1-SWSAP1-SPIDR drives the high level of sister-chromatid exchange, promotes long-range loss of heterozygosity often involved with cancer initiation, and impels the poor growth of BLM helicase-deficient cells. The relevance of these genetic interactions is evident as SWSAP1 loss prolongs Blm-mutant embryo survival, suggesting a possible druggable target for the treatment of Bloom syndrome.


Subject(s)
DNA-Binding Proteins/metabolism , Homologous Recombination/genetics , Multiprotein Complexes/metabolism , Animals , Bloom Syndrome/genetics , Bloom Syndrome/pathology , Cell Proliferation , HEK293 Cells , Humans , Meiosis , Mice , Mitosis , Mouse Embryonic Stem Cells/metabolism , Mutation/genetics , Phenotype , Rad51 Recombinase/metabolism , Sister Chromatid Exchange , Survival Analysis
6.
Article in English | MEDLINE | ID: mdl-33832920

ABSTRACT

Bloom syndrome is a rare autosomal recessive disorder with less than 300 cases reported in the literature. Bloom syndrome is characterized by chromosome instability, physical stigmata, growth deficiency, immunodeficiency, and a predisposition to cancer, most commonly leukemias, although solid tumors are reported as well. Bloom syndrome occurs in multiple ethnic groups with a higher incidence in persons of Ashkenazi Jewish origin. Few patients of Hispanic ethnicity have been reported. We report here a Mexican American family with a BLM pathogenic variant, c.2506_2507delAG, previously reported in a single patient from Mexico. In this family of four siblings, three have phenotypic features of Bloom syndrome, and BLM gene mutation was homozygous in these affected individuals. Our proband developed a rhabdomyosarcoma. Analysis of surrounding markers in the germline DNA revealed a common haplotype, suggesting a previously unrecognized founder mutation in the Hispanic population of Mexican origin.


Subject(s)
Bloom Syndrome/genetics , Mexican Americans , Mutation , Rhabdomyosarcoma/complications , Rhabdomyosarcoma/genetics , Alleles , Bloom Syndrome/pathology , Child, Preschool , Genetic Predisposition to Disease/genetics , Homozygote , Humans , Male , Mexico/epidemiology , Pedigree , Polymorphism, Single Nucleotide , RecQ Helicases/genetics , Rhabdomyosarcoma/pathology
7.
Mol Genet Metab ; 133(1): 35-48, 2021 05.
Article in English | MEDLINE | ID: mdl-33736941

ABSTRACT

Autosomal hereditary recessive diseases characterized by genetic instability are often associated with cancer predisposition. Bloom syndrome (BS), a rare genetic disorder, with <300 cases reported worldwide, combines both. Indeed, patients with Bloom's syndrome are 150 to 300 times more likely to develop cancers than normal individuals. The wide spectrum of cancers developed by BS patients suggests that early initial events occur in BS cells which may also be involved in the initiation of carcinogenesis in the general population and these may be common to several cancers. BS is caused by mutations of both copies of the BLM gene, encoding the RecQ BLM helicase. This review discusses the different aspects of BS and the different cellular functions of BLM in genome surveillance and maintenance through its major roles during DNA replication, repair, and transcription. BLM's activities are essential for the stabilization of centromeric, telomeric and ribosomal DNA sequences, and the regulation of innate immunity. One of the key objectives of this work is to establish a link between BLM functions and the main clinical phenotypes observed in BS patients, as well as to shed new light on the correlation between the genetic instability and diseases such as immunodeficiency and cancer. The different potential implications of the BLM helicase in the tumorigenic process and the use of BLM as new potential target in the field of cancer treatment are also debated.


Subject(s)
Bloom Syndrome/genetics , Neoplasms/genetics , RecQ Helicases/genetics , Bloom Syndrome/complications , Bloom Syndrome/pathology , DNA Replication/genetics , Genomic Instability , Humans , Mutation/genetics , Neoplasms/complications , Neoplasms/pathology , Protein Binding/genetics
8.
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
9.
Sci Rep ; 11(1): 2157, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33495511

ABSTRACT

Bloom Syndrome (BS; OMIM #210900; ORPHA #125) is a rare genetic disorder that is associated with growth deficits, compromised immune system, insulin resistance, genome instability and extraordinary predisposition to cancer. Most efforts thus far have focused on understanding the role of the Bloom syndrome DNA helicase BLM as a recombination factor in maintaining genome stability and suppressing cancer. Here, we observed increased levels of reactive oxygen species (ROS) and DNA base damage in BLM-deficient cells, as well as oxidative-stress-dependent reduction in DNA replication speed. BLM-deficient cells exhibited increased mitochondrial mass, upregulation of mitochondrial transcription factor A (TFAM), higher ATP levels and increased respiratory reserve capacity. Cyclin B1, which acts in complex with cyclin-dependent kinase CDK1 to regulate mitotic entry and associated mitochondrial fission by phosphorylating mitochondrial fission protein Drp1, fails to be fully degraded in BLM-deficient cells and shows unscheduled expression in G1 phase cells. This failure to degrade cyclin B1 is accompanied by increased levels and persistent activation of Drp1 throughout mitosis and into G1 phase as well as mitochondrial fragmentation. This study identifies mitochondria-associated abnormalities in Bloom syndrome patient-derived and BLM-knockout cells and we discuss how these abnormalities may contribute to Bloom syndrome.


Subject(s)
Bloom Syndrome/enzymology , Bloom Syndrome/pathology , Mitochondria/metabolism , Oxidative Stress , RecQ Helicases/deficiency , Autophagy , Cyclin B1/metabolism , DNA Damage , DNA Replication , DNA-Binding Proteins/metabolism , Energy Metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , G1 Phase , Humans , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Mitosis , Reactive Oxygen Species/metabolism , RecQ Helicases/metabolism , Transcription Factors/metabolism , Up-Regulation
10.
Pediatr Blood Cancer ; 68(2): e28815, 2021 02.
Article in English | MEDLINE | ID: mdl-33226170

ABSTRACT

The treatment of malignancy in cancer predisposition syndromes that also confer exquisite sensitivity to standard chemotherapy and radiation regimens remains a challenge. Bloom syndrome is one such disorder that is caused by a defect in DNA repair, predisposing to the development of early-onset age-related medical conditions and malignancies. We report on two patients with Bloom syndrome who responded well to chemotherapy despite significant alterations to standard protocols necessitated by hypersensitivity. Both patients experienced severe toxicities and exacerbation of endocrine comorbidities during chemotherapy. A multidisciplinary team of oncologists and endocrinologists is best suited to care for this patient population.


Subject(s)
Antineoplastic Agents/therapeutic use , Bloom Syndrome/pathology , Endocrine System Diseases/pathology , Neoplasms/drug therapy , Antineoplastic Agents/adverse effects , Bloom Syndrome/genetics , DNA Repair/genetics , Female , Humans , Male , Neoplasms/pathology , RecQ Helicases/genetics , Young Adult
11.
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
12.
J Exp Med ; 216(5): 1199-1213, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30936263

ABSTRACT

Cellular innate immune sensors of DNA are essential for host defense against invading pathogens. However, the presence of self-DNA inside cells poses a risk of triggering unchecked immune responses. The mechanisms limiting induction of inflammation by self-DNA are poorly understood. BLM RecQ-like helicase is essential for genome integrity and is deficient in Bloom syndrome (BS), a rare genetic disease characterized by genome instability, accumulation of micronuclei, susceptibility to cancer, and immunodeficiency. Here, we show that BLM-deficient fibroblasts show constitutive up-regulation of inflammatory interferon-stimulated gene (ISG) expression, which is mediated by the cGAS-STING-IRF3 cytosolic DNA-sensing pathway. Increased DNA damage or down-regulation of the cytoplasmic exonuclease TREX1 enhances ISG expression in BLM-deficient fibroblasts. cGAS-containing cytoplasmic micronuclei are increased in BS cells. Finally, BS patients demonstrate elevated ISG expression in peripheral blood. These results reveal that BLM limits ISG induction, thus connecting DNA damage to cellular innate immune response, which may contribute to human pathogenesis.


Subject(s)
Immunity, Innate/immunology , Nucleotidyltransferases/metabolism , RecQ Helicases/immunology , 2',5'-Oligoadenylate Synthetase/genetics , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Bloom Syndrome/pathology , Child , Cytosol/metabolism , DNA Damage/immunology , Exodeoxyribonucleases/metabolism , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Humans , Interferon Regulatory Factor-3/metabolism , Male , Membrane Proteins/metabolism , Nucleotidyltransferases/genetics , Phosphoproteins/metabolism , RNA-Binding Proteins/genetics , RecQ Helicases/genetics , Transcriptome , Transduction, Genetic , Tumor Suppressor Proteins/genetics
13.
Nucleic Acids Res ; 47(9): 4694-4706, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30916344

ABSTRACT

DNA helicases of the RecQ family are conserved among the three domains of life and play essential roles in genome maintenance. Mutations in several human RecQ helicases lead to diseases that are marked by cancer predisposition. The Saccharomyces cerevisiae RecQ helicase Sgs1 is orthologous to human BLM, defects in which cause the cancer-prone Bloom's Syndrome. Here, we use single-molecule imaging to provide a quantitative mechanistic understanding of Sgs1 activities on single stranded DNA (ssDNA), which is a central intermediate in all aspects of DNA metabolism. We show that Sgs1 acts upon ssDNA bound by either replication protein A (RPA) or the recombinase Rad51. Surprisingly, we find that Sgs1 utilizes a novel motor mechanism for disrupting ssDNA intermediates bound by the recombinase protein Rad51. The ability of Sgs1 to disrupt Rad51-ssDNA filaments may explain some of the defects engendered by RECQ helicase deficiencies in human cells.


Subject(s)
Rad51 Recombinase/genetics , RecQ Helicases/genetics , Recombination, Genetic , Saccharomyces cerevisiae Proteins/genetics , Adenosine Triphosphate/genetics , Bloom Syndrome/genetics , Bloom Syndrome/pathology , DNA Repair/genetics , DNA, Single-Stranded , Humans , Mutation/genetics , Saccharomyces cerevisiae/genetics
14.
Eur J Med Genet ; 61(2): 94-97, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29056561

ABSTRACT

Bloom syndrome is an autosomal recessive condition characterized by severe pre- and postnatal growth deficiency, immunodeficiency, an increased risk for malignancies, craniofacial dysmorphisms, and "typical" erythematous sun-sensitive skin lesions of the face. This facial rash has a butterfly-shaped distribution around the nose and is usually observed for the first time during the early years of life. Though reported as being a main feature of Bloom syndrome, there seems to be phenotypic variability regarding this facial skin rash among patients. It has been previously reported that in some individuals with Bloom syndrome these sun-sensitive lesions are less prominent or even absent. In this report we describe a 36 year old woman with short stature, microcephaly, several dysmorphisms, congenital hypothyroidism and premature ovarian failure. She was diagnosed with nasopharyngeal carcinoma at 36 years of age, only a few months after her consultation at the department of Clinical Genetics. Whole Exome Sequencing demonstrated that she had Bloom syndrome caused by a compound heterozygous mutation in BLM (c.2207_2212delinsTAGATTC; p.(Tyr736Leufs*5) and c.3681del; p.(Lys1227Asnfs*52)). She did not have facial sun-sensitive erythematous rash during childhood nor adulthood. We conclude that Bloom syndrome does not always present with erythematous sun-sensitive skin lesions of the face. We would like to underline that phenotypic variation regarding this "hallmark" feature of Bloom syndrome exists. Being aware of this might prevent a delay in diagnosing this rare short-stature syndrome and, subsequently, its potential clinical implications.


Subject(s)
Bloom Syndrome/pathology , Erythema/pathology , Phenotype , Adult , Bloom Syndrome/genetics , Diagnosis, Differential , Erythema/etiology , Erythema/genetics , Female , Humans , RecQ Helicases/genetics , Sunlight/adverse effects
15.
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
16.
Nat Commun ; 8(1): 693, 2017 09 25.
Article in English | MEDLINE | ID: mdl-28947735

ABSTRACT

Cells from Bloom's syndrome patients display genome instability due to a defective BLM and the downregulation of cytidine deaminase. Here, we use a genome-wide RNAi-synthetic lethal screen and transcriptomic profiling to identify genes enabling BLM-deficient and/or cytidine deaminase-deficient cells to tolerate constitutive DNA damage and replication stress. We found a synthetic lethal interaction between cytidine deaminase and microtubule-associated protein Tau deficiencies. Tau is overexpressed in cytidine deaminase-deficient cells, and its depletion worsens genome instability, compromising cell survival. Tau is recruited, along with upstream-binding factor, to ribosomal DNA loci. Tau downregulation decreases upstream binding factor recruitment, ribosomal RNA synthesis, ribonucleotide levels, and affects ribosomal DNA stability, leading to the formation of a new subclass of human ribosomal ultrafine anaphase bridges. We describe here Tau functions in maintaining survival of cytidine deaminase-deficient cells, and ribosomal DNA transcription and stability. Moreover, our findings for cancer tissues presenting concomitant cytidine deaminase underexpression and Tau upregulation open up new possibilities for anti-cancer treatment.Cytidine deaminase (CDA) deficiency leads to genome instability. Here the authors find a synthetic lethal interaction between CDA and the microtubule-associated protein Tau deficiencies, and report that Tau depletion affects rRNA synthesis, ribonucleotide pool balance, and rDNA stability.


Subject(s)
Bloom Syndrome/genetics , DNA, Ribosomal/metabolism , tau Proteins/physiology , Bloom Syndrome/pathology , Cell Survival , Cytidine Deaminase/deficiency , Down-Regulation , Genomic Instability , HeLa Cells , Humans , RecQ Helicases/genetics , Up-Regulation , tau Proteins/genetics , tau Proteins/metabolism
17.
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
18.
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
19.
PLoS Genet ; 12(12): e1006483, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27977684

ABSTRACT

Bloom syndrome is a recessive human genetic disorder with features of genome instability, growth deficiency and predisposition to cancer. The only known causative gene is the BLM helicase that is a member of a protein complex along with topoisomerase III alpha, RMI1 and 2, which maintains replication fork stability and dissolves double Holliday junctions to prevent genome instability. Here we report the identification of a second gene, RMI2, that is deleted in affected siblings with Bloom-like features. Cells from homozygous individuals exhibit elevated rates of sister chromatid exchange, anaphase DNA bridges and micronuclei. Similar genome and chromosome instability phenotypes are observed in independently derived RMI2 knockout cells. In both patient and knockout cell lines reduced localisation of BLM to ultra fine DNA bridges and FANCD2 at foci linking bridges are observed. Overall, loss of RMI2 produces a partially active BLM complex with mild features of Bloom syndrome.


Subject(s)
Bloom Syndrome/genetics , DNA-Binding Proteins/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Neoplasms/genetics , Nuclear Proteins/genetics , Bloom Syndrome/complications , Bloom Syndrome/pathology , Chromosomal Instability/genetics , DNA Helicases/genetics , DNA, Cruciform/genetics , Genetic Predisposition to Disease , Genomic Instability , Humans , Multiprotein Complexes/genetics , Neoplasms/complications , Neoplasms/pathology , Sister Chromatid Exchange/genetics
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...