Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Mol Cell ; 84(9): 1684-1698.e9, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38593805

RESUMO

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.


Assuntos
Dano ao DNA , Replicação do DNA , RecQ Helicases , Homeostase do Telômero , Telômero , RecQ Helicases/metabolismo , RecQ Helicases/genética , Humanos , Telômero/metabolismo , Telômero/genética , DNA de Cadeia Simples/metabolismo , DNA de Cadeia Simples/genética , Proteína Nuclear Ligada ao X/genética , Proteína Nuclear Ligada ao X/metabolismo , DNA Helicases/metabolismo , DNA Helicases/genética , Síndrome de Bloom/genética , Síndrome de Bloom/metabolismo , Síndrome de Bloom/enzimologia , Síndrome de Bloom/patologia , Linhagem Celular Tumoral
2.
Mol Med Rep ; 27(5)2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37052241

RESUMO

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.


Assuntos
Síndrome de Bloom , Humanos , Criança , Lactente , Feminino , Masculino , Síndrome de Bloom/diagnóstico , Síndrome de Bloom/genética , Síndrome de Bloom/patologia , Sequenciamento do Exoma , Cromossomos Humanos Y , Mosaicismo , Heterozigoto
5.
Nat Commun ; 12(1): 4255, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34253720

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Recombinação Homóloga/genética , Complexos Multiproteicos/metabolismo , Animais , Síndrome de Bloom/genética , Síndrome de Bloom/patologia , Proliferação de Células , Células HEK293 , Humanos , Meiose , Camundongos , Mitose , Células-Tronco Embrionárias Murinas/metabolismo , Mutação/genética , Fenótipo , Rad51 Recombinase/metabolismo , Troca de Cromátide Irmã , Análise de Sobrevida
6.
Artigo em Inglês | MEDLINE | ID: mdl-33832920

RESUMO

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.


Assuntos
Síndrome de Bloom/genética , Americanos Mexicanos , Mutação , Rabdomiossarcoma/complicações , Rabdomiossarcoma/genética , Alelos , Síndrome de Bloom/patologia , Pré-Escolar , Predisposição Genética para Doença/genética , Homozigoto , Humanos , Masculino , México/epidemiologia , Linhagem , Polimorfismo de Nucleotídeo Único , RecQ Helicases/genética , Rabdomiossarcoma/patologia
7.
Mol Genet Metab ; 133(1): 35-48, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33736941

RESUMO

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.


Assuntos
Síndrome de Bloom/genética , Neoplasias/genética , RecQ Helicases/genética , Síndrome de Bloom/complicações , Síndrome de Bloom/patologia , Replicação do DNA/genética , Instabilidade Genômica , Humanos , Mutação/genética , Neoplasias/complicações , Neoplasias/patologia , Ligação Proteica/genética
8.
Sci Rep ; 11(1): 2157, 2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33495511

RESUMO

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.


Assuntos
Síndrome de Bloom/enzimologia , Síndrome de Bloom/patologia , Mitocôndrias/metabolismo , Estresse Oxidativo , RecQ Helicases/deficiência , Autofagia , Ciclina B1/metabolismo , Dano ao DNA , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Metabolismo Energético , Fibroblastos/enzimologia , Fibroblastos/patologia , Fase G1 , Humanos , Mitocôndrias/ultraestrutura , Proteínas Mitocondriais/metabolismo , Mitose , Espécies Reativas de Oxigênio/metabolismo , RecQ Helicases/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima
9.
Pediatr Blood Cancer ; 68(2): e28815, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33226170

RESUMO

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.


Assuntos
Antineoplásicos/uso terapêutico , Síndrome de Bloom/patologia , Doenças do Sistema Endócrino/patologia , Neoplasias/tratamento farmacológico , Antineoplásicos/efeitos adversos , Síndrome de Bloom/genética , Reparo do DNA/genética , Feminino , Humanos , Masculino , Neoplasias/patologia , RecQ Helicases/genética , Adulto Jovem
10.
J Exp Med ; 216(5): 1199-1213, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-30936263

RESUMO

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.


Assuntos
Imunidade Inata/imunologia , Nucleotidiltransferases/metabolismo , RecQ Helicases/imunologia , 2',5'-Oligoadenilato Sintetase/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Síndrome de Bloom/patologia , Criança , Citosol/metabolismo , Dano ao DNA/imunologia , Exodesoxirribonucleases/metabolismo , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Fator Regulador 3 de Interferon/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/genética , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/genética , RecQ Helicases/genética , Transcriptoma , Transdução Genética , Proteínas Supressoras de Tumor/genética
11.
Nucleic Acids Res ; 47(9): 4694-4706, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-30916344

RESUMO

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.


Assuntos
Rad51 Recombinase/genética , RecQ Helicases/genética , Recombinação Genética , Proteínas de Saccharomyces cerevisiae/genética , Trifosfato de Adenosina/genética , Síndrome de Bloom/genética , Síndrome de Bloom/patologia , Reparo do DNA/genética , DNA de Cadeia Simples , Humanos , Mutação/genética , Saccharomyces cerevisiae/genética
12.
Eur J Med Genet ; 61(2): 94-97, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29056561

RESUMO

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.


Assuntos
Síndrome de Bloom/patologia , Eritema/patologia , Fenótipo , Adulto , Síndrome de Bloom/genética , Diagnóstico Diferencial , Eritema/etiologia , Eritema/genética , Feminino , Humanos , RecQ Helicases/genética , Luz Solar/efeitos adversos
13.
J Cell Biol ; 216(12): 3991-4005, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29042409

RESUMO

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.


Assuntos
Síndrome de Bloom/genética , DNA/química , Instabilidade Genômica , RecQ Helicases/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Síndrome de Bloom/metabolismo , Síndrome de Bloom/patologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , DNA/genética , DNA/metabolismo , Reparo do DNA , Replicação do DNA , Fibroblastos/metabolismo , Fibroblastos/patologia , Dosagem de Genes , Regulação da Expressão Gênica , Histonas/genética , Histonas/metabolismo , Humanos , Conformação de Ácido Nucleico , Ligação Proteica , RNA/genética , RNA/metabolismo , RecQ Helicases/metabolismo , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/ultraestrutura , Proteínas de Saccharomyces cerevisiae/metabolismo
14.
Nat Commun ; 8(1): 693, 2017 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-28947735

RESUMO

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.


Assuntos
Síndrome de Bloom/genética , DNA Ribossômico/metabolismo , Proteínas tau/fisiologia , Síndrome de Bloom/patologia , Sobrevivência Celular , Citidina Desaminase/deficiência , Regulação para Baixo , Instabilidade Genômica , Células HeLa , Humanos , RecQ Helicases/genética , Regulação para Cima , Proteínas tau/genética , Proteínas tau/metabolismo
15.
Redox Biol ; 11: 375-383, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28063379

RESUMO

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.


Assuntos
Ataxia Telangiectasia/genética , Síndrome de Bloom/genética , Reparo do DNA , Mitocôndrias/metabolismo , Síndrome de Quebra de Nijmegen/genética , Estresse Oxidativo/genética , Ataxia Telangiectasia/metabolismo , Ataxia Telangiectasia/patologia , Síndrome de Bloom/metabolismo , Síndrome de Bloom/patologia , Dano ao DNA , Regulação da Expressão Gênica , Humanos , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Mitocôndrias/patologia , NADPH Oxidase 4/genética , NADPH Oxidase 4/metabolismo , Síndrome de Quebra de Nijmegen/metabolismo , Síndrome de Quebra de Nijmegen/patologia , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
16.
PLoS Genet ; 12(12): e1006483, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27977684

RESUMO

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.


Assuntos
Síndrome de Bloom/genética , Proteínas de Ligação a DNA/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Neoplasias/genética , Proteínas Nucleares/genética , Síndrome de Bloom/complicações , Síndrome de Bloom/patologia , Instabilidade Cromossômica/genética , DNA Helicases/genética , DNA Cruciforme/genética , Predisposição Genética para Doença , Instabilidade Genômica , Humanos , Complexos Multiproteicos/genética , Neoplasias/complicações , Neoplasias/patologia , Troca de Cromátide Irmã/genética
17.
Oncotarget ; 7(22): 32351-61, 2016 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-27083049

RESUMO

Mutations in the human RecQ helicase, BLM, causes Bloom Syndrome, which is a rare autosomal recessive disorder and characterized by genomic instability and an increased risk of cancer. Fanconi Anemia (FA), resulting from mutations in any of the 19 known FA genes and those yet to be known, is also characterized by chromosomal instability and a high incidence of cancer. BLM helicase and FA proteins, therefore, may work in a common tumor-suppressor signaling pathway. To date, it remains largely unclear as to how BLM and FA proteins work concurrently in the maintenance of genome stability. Here we report that BLM is involved in the early activation of FA group D2 protein (FANCD2). We found that FANCD2 activation is substantially delayed and attenuated in crosslinking agent-treated cells harboring deficient Blm compared to similarly treated control cells with sufficient BLM. We also identified that the domain VI of BLM plays an essential role in promoting FANCD2 activation in cells treated with DNA crosslinking agents, especially ultraviolet B. The similar biological effects performed by ΔVI-BLM and inactivated FANCD2 further confirm the relationship between BLM and FANCD2. Mutations within the domain VI of BLM detected in human cancer samples demonstrate the functional importance of this domain, suggesting human tumorigenicity resulting from mtBLM may be at least partly attributed to mitigated FANCD2 activation. Collectively, our data show a previously unknown regulatory liaison in advancing our understanding of how the cancer susceptibility gene products act in concert to maintain genome stability.


Assuntos
Síndrome de Bloom/enzimologia , Proteína do Grupo de Complementação L da Anemia de Fanconi/metabolismo , Anemia de Fanconi/enzimologia , Neoplasias/enzimologia , RecQ Helicases/metabolismo , Transdução de Sinais , Síndrome de Bloom/genética , Síndrome de Bloom/patologia , Neoplasias Ósseas/enzimologia , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Reagentes de Ligações Cruzadas/farmacologia , Anemia de Fanconi/genética , Anemia de Fanconi/patologia , Proteína do Grupo de Complementação L da Anemia de Fanconi/química , Proteína do Grupo de Complementação L da Anemia de Fanconi/genética , Feminino , Humanos , Mutação , Neoplasias/genética , Neoplasias/patologia , Osteossarcoma/enzimologia , Osteossarcoma/genética , Osteossarcoma/patologia , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Interferência de RNA , RecQ Helicases/química , RecQ Helicases/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Transfecção , Raios Ultravioleta
18.
DNA Repair (Amst) ; 41: 73-84, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27100209

RESUMO

Mutation of Bloom helicase (BLM) causes Bloom syndrome (BS), a rare human genetic disorder associated with genome instability, elevation of sister chromatid exchanges, and predisposition to cancer. Deficiency in BLM homologs in Drosophila and yeast brings about significantly increased rates of recombination between imperfectly matched sequences ("homeologous recombination," or HeR). To assess whether BLM deficiency provokes an increase in HeR in human cells, we transfected an HeR substrate into a BLM-null cell line derived from a BS patient. The substrate contained a thymidine kinase (tk)-neo fusion gene disrupted by the recognition site for endonuclease I-SceI, as well as a functional tk gene to serve as a potential recombination partner for the tk-neo gene. The two tk sequences on the substrate displayed 19% divergence. A double-strand break was introduced by expression of I-SceI and repair events were recovered by selection for G418-resistant clones. Among 181 events recovered, 30 were accomplished via HeR with the balance accomplished by nonhomologous end-joining. The frequency of HeR events in the BS cells was elevated significantly compared to that seen in normal human fibroblasts or in BS cells complemented for BLM expression. We conclude that BLM deficiency enables HeR in human cells.


Assuntos
Cromossomos Humanos/genética , Recombinação Homóloga , RecQ Helicases/deficiência , RecQ Helicases/genética , Sequência de Bases , Síndrome de Bloom/genética , Síndrome de Bloom/patologia , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Humanos , Mutação
19.
PLoS Genet ; 11(7): e1005384, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26181065

RESUMO

Genome stability is jeopardized by imbalances of the dNTP pool; such imbalances affect the rate of fork progression. For example, cytidine deaminase (CDA) deficiency leads to an excess of dCTP, slowing the replication fork. We describe here a novel mechanism by which pyrimidine pool disequilibrium compromises the completion of replication and chromosome segregation: the intracellular accumulation of dCTP inhibits PARP-1 activity. CDA deficiency results in incomplete DNA replication when cells enter mitosis, leading to the formation of ultrafine anaphase bridges between sister-chromatids at "difficult-to-replicate" sites such as centromeres and fragile sites. Using molecular combing, electron microscopy and a sensitive assay involving cell imaging to quantify steady-state PAR levels, we found that DNA replication was unsuccessful due to the partial inhibition of basal PARP-1 activity, rather than slower fork speed. The stimulation of PARP-1 activity in CDA-deficient cells restores replication and, thus, chromosome segregation. Moreover, increasing intracellular dCTP levels generates under-replication-induced sister-chromatid bridges as efficiently as PARP-1 knockdown. These results have direct implications for Bloom syndrome (BS), a rare genetic disease combining susceptibility to cancer and genomic instability. BS results from mutation of the BLM gene, encoding BLM, a RecQ 3'-5' DNA helicase, a deficiency of which leads to CDA downregulation. BS cells thus have a CDA defect, resulting in a high frequency of ultrafine anaphase bridges due entirely to dCTP-dependent PARP-1 inhibition and independent of BLM status. Our study describes previously unknown pathological consequences of the distortion of dNTP pools and reveals an unexpected role for PARP-1 in preventing DNA under-replication and chromosome segregation defects.


Assuntos
Síndrome de Bloom/genética , Citidina Desaminase/genética , Poli(ADP-Ribose) Polimerases/genética , Pirimidinas/metabolismo , Síndrome de Bloom/patologia , Linhagem Celular , Centrômero/genética , Sítios Frágeis do Cromossomo/genética , Segregação de Cromossomos/genética , Citidina Desaminase/deficiência , Replicação do DNA/genética , Instabilidade Genômica , Humanos , Mitose/genética , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/biossíntese , RecQ Helicases/genética , Troca de Cromátide Irmã/genética
20.
Pediatr Hematol Oncol ; 30(6): 544-53, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23647505

RESUMO

BACKGROUND: An increased incidence of non-Hodgkin lymphoma (NHL) has been seen in various primary immune deficiency (PID) cases. The present study aimed to evaluate the clinical characteristics and treatment outcomes of five cases with NHL associated with primary immunodeficiency. METHODS: We retrospectively evaluated five patients with primary immunodeficiency who developed NHL. Two patients had ataxia-telangiectasia (A-T), one patient had common variable immunodeficiency (CVID), one patient had Bloom's Syndrome, and one patient had Wiskott-Aldrich syndrome (WAS). RESULTS: All patients were male (median age, 8 years). Stage distribution was stage III in three patients and stage IV in two patients. Three patients had B-cell lymphoma and two had T-cell lymphoma. Reduced doses of Berlin-Frankfurt-Münster (BFM) and French Society of Pediatric Oncology (SFOP) regimens were used in four patients according to histopathological subtype. The two patients with ataxia and one patient with Bloom's Syndrome died of progressive/relapsed disease at months 5, 19, and 6, respectively. The patient with CVID associated with T-cell lymphoma has been in remission for 7 years. A full-dosage regimen of rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) was successfully used in the patient with WAS and B-cell lymphoma; he was still in remission after 3 years. CONCLUSION: Primary immunodeficiency diseases are one of the strongest known risk factors for the development of NHL. Management of these patients remains problematic. There is a great need to develop new therapeutic approaches in this group. The use of rituximab in combination with CHOP may provide a promising treatment option for B-cell lymphomas associated with immunodeficiency.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Imunodeficiência de Variável Comum , Linfoma não Hodgkin , Síndrome de Wiskott-Aldrich , Adolescente , Adulto , Anticorpos Monoclonais Murinos/administração & dosagem , Ataxia Telangiectasia/complicações , Ataxia Telangiectasia/tratamento farmacológico , Ataxia Telangiectasia/patologia , Síndrome de Bloom/complicações , Síndrome de Bloom/tratamento farmacológico , Síndrome de Bloom/patologia , Criança , Imunodeficiência de Variável Comum/complicações , Imunodeficiência de Variável Comum/tratamento farmacológico , Imunodeficiência de Variável Comum/patologia , Ciclofosfamida/administração & dosagem , Doxorrubicina/administração & dosagem , Feminino , Humanos , Lactente , Recém-Nascido , Linfoma não Hodgkin/tratamento farmacológico , Linfoma não Hodgkin/etiologia , Linfoma não Hodgkin/patologia , Masculino , Prednisona/administração & dosagem , Estudos Retrospectivos , Fatores de Risco , Rituximab , Vincristina/administração & dosagem , Síndrome de Wiskott-Aldrich/complicações , Síndrome de Wiskott-Aldrich/tratamento farmacológico , Síndrome de Wiskott-Aldrich/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA