Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Rep (Hoboken) ; 6(2): e1700, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36806726

RESUMEN

BACKGROUND: Nijmegen breakage syndrome (NBS) is an autosomal-recessive chromosome instability disorder characterized by, among others, hypersensitivity to X-irradiation and an exceptionally high risk for lymphoid malignancy. The vast majority of NBS patients is homozygous for a common Slavic founder mutation, c.657del5, of the NBN gene, which is involved in the repair of DNA double-strand breaks (DSBs). The founder mutation also predisposes heterozygous carriers to cancer, apparently however, with a higher risk in the Czech Republic/Slovakia (CS) than in Poland. AIM: To examine whether the age of cancer manifestation and cancer death of NBN homozygotes is different between probands from CS and Poland. METHODS: The study is restricted to probands born until 1989, before replacement of the communist regime by a democratic system in CS and Poland, and a substantial transition of the health care systems. Moreover, all patients were recruited without knowledge of their genetic status since the NBN gene was not identified until 1998. RESULTS: Here, we show that cancer manifestation of NBN homozygotes is at a significantly earlier age in probands from CS than from Poland. This is explained by the difference in natural and medical radiation exposure, though within the permissible dosage. CONCLUSION: It is reasonable to assume that this finding also sheds light on the higher cancer risk of NBN heterozygotes in CS than in Poland. This has implications for genetic counseling and individualized medicine also of probands with other DNA repair defects.


Asunto(s)
Neoplasias , Síndrome de Nijmegen , Humanos , Proteínas Nucleares/genética , Proteínas de Ciclo Celular/genética , Heterocigoto , Síndrome de Nijmegen/genética , Síndrome de Nijmegen/patología , Mutación
2.
Hum Genet ; 141(11): 1785-1794, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35536377

RESUMEN

The evolutionary conserved Polo-like kinase 4 (PLK4) is essential for centriole duplication, spindle assembly, and de novo centriole formation. In man, homozygous mutations in PLK4 lead to primary microcephaly, altered PLK4 expression is associated with aneuploidy in human embryos. Here, we report on a consanguineous four-generation family with 8 affected individuals compound heterozygous for a novel missense variant, c.881 T > G, and a deletion of the PLK4 gene. The clinical phenotype of the adult patients is mild compared to individuals with previously described PLK4 mutations. One individual was homozygous for the variant c.881G and phenotypically unaffected. The deletion was inherited by 14 of 16 offspring and thus exhibits transmission ratio distortion (TRD). Moreover, based on the already published families with PLK4 mutations, it could be shown that due to the preferential transmission of the mutant alleles, the number of affected offspring is significantly increased. It is assumed that reduced expression of PLK4 decreases the intrinsically high error rate of the first cell divisions after fertilization, increases the number of viable embryos and thus leads to preferential transmission of the deleted/mutated alleles.


Asunto(s)
Proteínas de Ciclo Celular , Centriolos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , División Celular , Centriolos/genética , Centriolos/metabolismo , Humanos , Mutación , Proteínas Serina-Treonina Quinasas/genética
3.
Aging (Albany NY) ; 12(12): 12342-12375, 2020 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-32564008

RESUMEN

BACKGROUND: Nibrin, as part of the NBN/MRE11/RAD50 complex, is mutated in Nijmegen breakage syndrome (NBS), which leads to impaired DNA damage response and lymphoid malignancy. RESULTS: Telomere length (TL) was markedly reduced in homozygous patients (and comparably so in all chromosomes) by ~40% (qPCR) and was slightly reduced in NBS heterozygotes older than 30 years (~25% in qPCR), in accordance with the respective cancer rates. Humanized cancer-free NBS mice had normal TL. Telomere elongation was inducible by telomerase and/or alternative telomere lengthening but was associated with abnormal expression of telomeric genes involved in aging and/or cell growth. Lymphoblastoid cells from NBS patients with long survival times (>12 years) displayed the shortest telomeres and low caspase 7 activity. CONCLUSIONS: NBS is a secondary telomeropathy. The two-edged sword of telomere attrition enhances the cancer-prone situation in NBS but can also lead to a relatively stable cellular phenotype in tumor survivors. Results suggest a modular model for progeroid syndromes with abnormal expression of telomeric genes as a molecular basis. METHODS: We studied TL and function in 38 homozygous individuals, 27 heterozygotes, one homozygous fetus, six NBS lymphoblastoid cell lines, and humanized NBS mice, all with the same founder NBN mutation: c.657_661del5.


Asunto(s)
Proteínas de Ciclo Celular/genética , Síndrome de Nijmegen/complicaciones , Proteínas Nucleares/genética , Progeria/genética , Homeostasis del Telómero/genética , Telómero/patología , Adolescente , Animales , Línea Celular Tumoral , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Heterocigoto , Homocigoto , Humanos , Lactante , Cariotipificación , Masculino , Ratones , Ratones Transgénicos , Síndrome de Nijmegen/genética , Síndrome de Nijmegen/patología , Progeria/patología , Telomerasa/metabolismo , Adulto Joven
4.
PLoS One ; 13(11): e0207315, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30440001

RESUMEN

The genes, XRS2 in Saccharomyces cerevisiae and NBN in mammals, have little sequence identity at the amino acid level. Nevertheless, they are both found together with MRE11 and RAD50 in a highly conserved protein complex which functions in the repair of DNA double-strand breaks. Here, we have examined the evolutionary and functional relationship of these two genes by cross-complementation experiments. These experiments necessitated sequence correction for specific codon usage before they could be successfully conducted. We present evidence that despite extreme sequence divergence nibrin can, at least partially, replace Xrs2 in the cellular DNA damage response, and Xrs2 is able to promote nuclear localization of MRE11 in NBS cells. We discuss that the extreme sequence divergence reflects a unique adaptive pressure during evolution related to the specific eukaryotic role for both Xrs2 and nibrin in the subcellular localisation of the DNA repair complex. This, we suggest, is of particular relevance when cells are infected by viruses. The conflict hypothesis of co-evolution of DNA repair genes and DNA viruses may thus explain the very low sequence identity of these two homologous genes.


Asunto(s)
Proteínas de Ciclo Celular , Codón , Daño del ADN , Prueba de Complementación Genética , Proteínas Nucleares , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Transformada , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
5.
J Clin Invest ; 127(5): 1700-1713, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28346228

RESUMEN

It is well established that somatic genomic changes can influence phenotypes in cancer, but the role of adaptive changes in developmental disorders is less well understood. Here we have used next-generation sequencing approaches to identify de novo heterozygous mutations in sterile α motif domain-containing protein 9 (SAMD9, located on chromosome 7q21.2) in 8 children with a multisystem disorder termed MIRAGE syndrome that is characterized by intrauterine growth restriction (IUGR) with gonadal, adrenal, and bone marrow failure, predisposition to infections, and high mortality. These mutations result in gain of function of the growth repressor product SAMD9. Progressive loss of mutated SAMD9 through the development of monosomy 7 (-7), deletions of 7q (7q-), and secondary somatic loss-of-function (nonsense and frameshift) mutations in SAMD9 rescued the growth-restricting effects of mutant SAMD9 proteins in bone marrow and was associated with increased length of survival. However, 2 patients with -7 and 7q- developed myelodysplastic syndrome, most likely due to haploinsufficiency of related 7q21.2 genes. Taken together, these findings provide strong evidence that progressive somatic changes can occur in specific tissues and can subsequently modify disease phenotype and influence survival. Such tissue-specific adaptability may be a more common mechanism modifying the expression of human genetic conditions than is currently recognized.


Asunto(s)
Insuficiencia Suprarrenal/congénito , Deleción Cromosómica , Mutación del Sistema de Lectura , Haploinsuficiencia , Síndromes Mielodisplásicos/genética , Proteínas/genética , Insuficiencia Suprarrenal/genética , Insuficiencia Suprarrenal/mortalidad , Cromosomas Humanos Par 7 , Estudios de Cohortes , Mutación del Sistema de Lectura/genética , Humanos , Lactante , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular , Masculino , Síndromes Mielodisplásicos/mortalidad
6.
PLoS One ; 11(12): e0167984, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27936167

RESUMEN

The vast majority of patients with Nijmegen Breakage Syndrome (NBS) are of Slavic origin and carry a deleterious deletion (c.657del5; rs587776650) in the NBN gene on chromosome 8q21. This mutation is essentially confined to Slavic populations and may thus be considered a Slavic founder mutation. Notably, not a single parenthood of a homozygous c.657del5 carrier has been reported to date, while heterozygous carriers do reproduce but have an increased cancer risk. These observations seem to conflict with the considerable carrier frequency of c.657del5 of 0.5% to 1% as observed in different Slavic populations because deleterious mutations would be eliminated quite rapidly by purifying selection. Therefore, we propose that heterozygous c.657del5 carriers have increased reproductive success, i.e., that the mutation confers heterozygote advantage. In fact, in our cohort study of the reproductive history of 24 NBS pedigrees from the Czech Republic, we observed that female carriers gave birth to more children on average than female non-carriers, while no such reproductive differences were observed for males. We also estimate that c.657del5 likely occurred less than 300 generations ago, thus supporting the view that the original mutation predated the historic split and subsequent spread of the 'Slavic people'. We surmise that the higher fertility of female c.657del5 carriers reflects a lower miscarriage rate in these women, thereby reflecting the role of the NBN gene product, nibrin, in the repair of DNA double strand breaks and their processing in immune gene rearrangements, telomere maintenance, and meiotic recombination, akin to the previously described role of the DNA repair genes BRCA1 and BRCA2.


Asunto(s)
Proteínas de Ciclo Celular/genética , Efecto Fundador , Mutación , Síndrome de Nijmegen/genética , Proteínas Nucleares/genética , Reproducción/genética , Adulto , Estudios de Cohortes , República Checa , Daño del ADN , Reparación del ADN , Femenino , Tamización de Portadores Genéticos , Haplotipos , Humanos , Masculino , Persona de Mediana Edad , Síndrome de Nijmegen/etnología , Eslovaquia
7.
Mol Ther ; 24(1): 117-24, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26265251

RESUMEN

Over 90% of patients with Nijmegen breakage syndrome (NBS), a hereditary cancer disorder, are homoallelic for a 5 bp deletion in the NBN gene involved in the cellular response to DNA damage. This hypomorphic mutation leads to a carboxy-terminal protein fragment, p70-nibrin, with some residual function. Average age at malignancy, typically lymphoma, is 9.7 years. NBS patients are hypersensitive to chemotherapeutic and radiotherapeutic treatments, thus prevention of cancer development is of particular importance. Expression of an internally deleted NBN protein, p80-nibrin, has been previously shown to be associated with a milder cellular phenotype and absence of cancer in a 62-year-old NBS patient. Here we show that cells from this patient, unlike other NBS patients, have DNA replication and origin firing rates comparable to control cells. We used here antisense oligonucleotides to enforce alternative splicing in NBS patient cells and efficiently generate the same internally deleted p80-nibrin protein. Injecting the same antisense sequences as morpholino oligomers (VivoMorpholinos) into the tail vein of a humanized NBS murine mouse model also led to efficient alternative splicing in vivo. Thus, proof of principle for the use of antisense oligonucleotides as a potential cancer prophylaxis has been demonstrated.


Asunto(s)
Empalme Alternativo , Proteínas de Ciclo Celular/genética , Síndrome de Nijmegen/terapia , Proteínas Nucleares/genética , Oligonucleótidos Antisentido/administración & dosificación , Eliminación de Secuencia , Empalme Alternativo/efectos de los fármacos , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular , Niño , Replicación del ADN , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Persona de Mediana Edad , Síndrome de Nijmegen/genética , Proteínas Nucleares/antagonistas & inhibidores , Oligonucleótidos Antisentido/farmacología
8.
Mutat Res ; 769: 11-6, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25771721

RESUMEN

Nibrin (NBN) is a member of a DNA repair complex together with MRE11 and RAD50. The complex is associated particularly with the repair of DNA double strand breaks and with the regulation of cell cycle check points. Hypomorphic mutation of components of the complex leads to human disorders characterised by radiosensitivity and increased tumour occurrence, particularly of the lymphatic system. We have examined here the relationship between DNA damage, mutation frequency and mutation spectrum in vitro and in vivo in mouse models carrying NBN mutations and a lacZ reporter plasmid. We find that NBN mutation leads to increased spontaneous DNA damage in fibroblasts in vitro and high basal mutation rates in lymphatic tissue of mice in vivo. The characteristic mutation spectrum is dominated by single base transitions rather than the deletions and complex rearrangements expected after abortive repair of DNA double strand breaks. We conclude that in the absence of wild type nibrin, the repair of spontaneous errors, presumably arising during DNA replication, makes a major contribution to the basal mutation rate. This applies also to cells heterozygous for an NBN null mutation. Mutation frequencies after irradiation in vivo were not increased in mice with nibrin mutations as might have been expected considering the radiosensitivity of NBS patient cells in vitro. Evidently apoptosis is efficient, even in the absence of wild type nibrin.


Asunto(s)
Proteínas de Ciclo Celular/genética , Daño del ADN , Rayos gamma/efectos adversos , Tasa de Mutación , Proteínas Nucleares/genética , Animales , Proteínas de Ciclo Celular/deficiencia , Células Cultivadas , Rotura Cromosómica/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Daño del ADN/genética , Reparación del ADN/genética , Replicación del ADN/genética , Proteínas de Unión al ADN , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Nucleares/deficiencia
9.
Gene ; 527(2): 448-55, 2013 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-23860323

RESUMEN

Mutations in the fibrillin-1 gene (FBN1) cause Marfan Syndrome (MFS), a hereditary disorder of connective tissue. The transcription of FBN1 has been reported to be driven by a short ultraconserved region (SUPR) in the 5' untranslated exon A of FBN1, but the nature of other factors involved in FBN1 gene regulation has not been clarified. In this study, we characterized the transcription factors involved in FBN1 gene regulation. The results show that Sp1 protein binds to two putative binding sites in the promoter of FBN1. Overexpression of Sp1 resulted in a significant increase in both promoter activity and FBN1 mRNA level in HEK 293 cells, whereas inhibition or knockdown of Sp1 decreased FBN1 gene expression. In addition, we found that Poly [ADP-ribose] polymerase 1 (PARP1) binds to the palindromic sequence TCTCGCGAGA in the ultraconserved region of the FBN1 promoter and that the regulation of FBN1 expression by PARP1 is dependent on Sp1. These results indicate that both Sp1 and PARP1 contribute to FBN1 gene expression. These observations add to our understanding of the transcriptional regulation of FBN1 gene expression.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Proteínas de Microfilamentos/genética , Factor de Transcripción Sp1/fisiología , Secuencia de Bases , Cartilla de ADN , Fibrilina-1 , Fibrilinas , Células HEK293 , Humanos , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción Sp1/metabolismo
10.
Gene ; 519(2): 217-21, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23458873

RESUMEN

The autosomal recessive disorder Nijmegen breakage syndrome (NBS) is caused by mutations in the NBN gene which codes for the protein nibrin (NBS1; p95). In the majority of cases, a 5bp deletion, a founder mutation, leads to a hypomorphic 70kD protein, p70-nibrin, after alternative initiation of translation. Protein levels are of relevance for the clinical course of the disease, particularly with regard to malignancy. Here, mechanisms and efficiency of mutant protein clearance were examined in order to establish whether these have an impact on nibrin abundance. Cell lines from NBS patients and retroviral transductants were treated with proteasome and lysosome inhibitors and examined by semi-quantitative immunoblotting for p70-nibrin and p95-nibrin levels. The results show that p70-nibrin is degraded by the proteasome with varying efficiency in cell lines from different NBS patients leading to lower or higher steady state levels of this partially active protein fragment. In contrast, a previously described NBN missense mutation, which disturbs protein folding due to the substitution of a critical arginine by tryptophan, was found to be cleared by lysosomal microautophagy leading also to lower cellular levels. The data show that truncated nibrin and misfolded nibrin have different clearance pathways.


Asunto(s)
Alelos , Proteínas de Ciclo Celular/genética , Proteínas Mutantes/genética , Síndrome de Nijmegen/genética , Proteínas Nucleares/genética , Sustitución de Aminoácidos , Autofagia/genética , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Marcadores Genéticos , Vectores Genéticos , Humanos , Cinética , Mutación Missense , Síndrome de Nijmegen/patología , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Retroviridae/genética , Retroviridae/metabolismo
11.
Hum Mol Genet ; 21(22): 4948-56, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22907656

RESUMEN

The recessive genetic disorder Fanconi anemia (FA) is clinically characterized by congenital defects, bone marrow failure and an increased incidence of cancer. Cells derived from FA patients exhibit hypersensitivity to DNA interstrand crosslink (ICL)-inducing agents. We have earlier reported a similar cellular phenotype for human cells depleted of hSNM1B/Apollo (siRNA). In fact, hSNM1B/Apollo has a dual role in the DNA damage response and in generation and maintenance of telomeres, the latter function involving interaction with the shelterin protein TRF2. Here we find that ectopically expressed hSNM1B/Apollo co-immunoprecipitates with SLX4, a protein recently identified as a new FA protein, FANCP, and known to interact with several structure-specific nucleases. As shown by immunofluorescence analysis, FANCP/SLX4 depletion (siRNA) resulted in a significant reduction of hSNM1B/Apollo nuclear foci, supporting the functional relevance of this new protein interaction. Interestingly, as an additional consequence of FANCP/SLX4 depletion, we found a reduction of cellular TRF2, in line with its telomere-related function. Finally, analysis of human cells following double knockdown of hSNM1B/Apollo and FANCP/SLX4 indicated that they function epistatically. These findings further substantiate the role of hSNM1B/Apollo in a downstream step of the FA pathway during the repair of DNA ICLs.


Asunto(s)
Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Recombinasas/metabolismo , Transducción de Señal , Línea Celular , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Exodesoxirribonucleasas , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Silenciador del Gen , Humanos , Mitomicina/farmacología , Unión Proteica , Recombinasas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
12.
Orphanet J Rare Dis ; 7: 13, 2012 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-22373003

RESUMEN

Nijmegen breakage syndrome (NBS) is a rare autosomal recessive syndrome of chromosomal instability mainly characterized by microcephaly at birth, combined immunodeficiency and predisposition to malignancies. Due to a founder mutation in the underlying NBN gene (c.657_661del5) the disease is encountered most frequently among Slavic populations. The principal clinical manifestations of the syndrome are: microcephaly, present at birth and progressive with age, dysmorphic facial features, mild growth retardation, mild-to-moderate intellectual disability, and, in females, hypergonadotropic hypogonadism. Combined cellular and humoral immunodeficiency with recurrent sinopulmonary infections, a strong predisposition to develop malignancies (predominantly of lymphoid origin) and radiosensitivity are other integral manifestations of the syndrome. The NBN gene codes for nibrin which, as part of a DNA repair complex, plays a critical nuclear role wherever double-stranded DNA ends occur, either physiologically or as a result of mutagenic exposure. Laboratory findings include: (1) spontaneous chromosomal breakage in peripheral T lymphocytes with rearrangements preferentially involving chromosomes 7 and 14, (2) sensitivity to ionizing radiation or radiomimetics as demonstrated in vitro by cytogenetic methods or by colony survival assay, (3) radioresistant DNA synthesis, (4) biallelic hypomorphic mutations in the NBN gene, and (5) absence of full-length nibrin protein. Microcephaly and immunodeficiency are common to DNA ligase IV deficiency (LIG4 syndrome) and severe combined immunodeficiency with microcephaly, growth retardation, and sensitivity to ionizing radiation due to NHEJ1 deficiency (NHEJ1 syndrome). In fact, NBS was most commonly confused with Fanconi anaemia and LIG4 syndrome. Genetic counselling should inform parents of an affected child of the 25% risk for further children to be affected. Prenatal molecular genetic diagnosis is possible if disease-causing mutations in both alleles of the NBN gene are known. No specific therapy is available for NBS, however, hematopoietic stem cell transplantation may be one option for some patients. Prognosis is generally poor due to the extremely high rate of malignancies.


Asunto(s)
Síndrome de Nijmegen , Adolescente , Adulto , Proteínas de Ciclo Celular/genética , Preescolar , Inestabilidad Cromosómica/genética , Femenino , Genes Recesivos , Humanos , Hipogonadismo/genética , Hipogonadismo/patología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/fisiopatología , Microcefalia/genética , Microcefalia/patología , Neoplasias/genética , Neoplasias/patología , Síndrome de Nijmegen/complicaciones , Síndrome de Nijmegen/genética , Síndrome de Nijmegen/patología , Síndrome de Nijmegen/fisiopatología , Proteínas Nucleares/genética , Adulto Joven
13.
PLoS Genet ; 8(3): e1002557, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22396666

RESUMEN

Nijmegen Breakage Syndrome (NBS), an autosomal recessive genetic instability syndrome, is caused by hypomorphic mutation of the NBN gene, which codes for the protein nibrin. Nibrin is an integral member of the MRE11/RAD50/NBN (MRN) complex essential for processing DNA double-strand breaks. Cardinal features of NBS are immunodeficiency and an extremely high incidence of hematological malignancies. Recent studies in conditional null mutant mice have indicated disturbances in redox homeostasis due to impaired DSB processing. Clearly this could contribute to DNA damage, chromosomal instability, and cancer occurrence. Here we show, in the complete absence of nibrin in null mutant mouse cells, high levels of reactive oxygen species several hours after exposure to a mutagen. We show further that NBS patient cells, which unlike mouse null mutant cells have a truncated nibrin protein, also have high levels of reactive oxygen after DNA damage and that this increased oxidative stress is caused by depletion of NAD+ due to hyperactivation of the strand-break sensor, Poly(ADP-ribose) polymerase. Both hyperactivation of Poly(ADP-ribose) polymerase and increased ROS levels were reversed by use of a specific Poly(ADP-ribose) polymerase inhibitor. The extremely high incidence of malignancy among NBS patients is the result of the combination of a primary DSB repair deficiency with secondary oxidative DNA damage.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Síndrome de Nijmegen , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Bleomicina/farmacología , Células Cultivadas , Roturas del ADN de Doble Cadena , Daño del ADN/efectos de los fármacos , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Mutantes , NAD/metabolismo , Síndrome de Nijmegen/genética , Síndrome de Nijmegen/metabolismo , Estrés Oxidativo/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Especies Reactivas de Oxígeno/metabolismo , Activación Transcripcional
14.
Neurogenetics ; 12(4): 273-82, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21965147

RESUMEN

Ataxia telangiectasia (AT) is an autosomal recessive disorder characterized by cerebellar degeneration, immunodeficiency, oculocutaneous telangiectasias, chromosomal instability, radiosensitivity, and cancer predisposition. The gene mutated in the patients, ATM, encodes a member of the phosphatidylinositol 3-kinase family proteins. The ATM protein has a key role in the cellular response to DNA damage. Truncating and splice site mutations in ATM have been found in most patients with the classical AT phenotype. Here we report of our extensive ATM mutation screening on 25 AT patients from 19 families of different ethnic origin. Previously unknown mutations were identified in six patients including a new homozygous missense mutation, c.8110T>C (p.Cys2704Arg), in a severely affected patient. Comprehensive clinical data are presented for all patients described here along with data on ATM function generated by analysis of cell lines established from a subset of the patients.


Asunto(s)
Ataxia Telangiectasia/genética , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Mutación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/genética , Adolescente , Adulto , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Niño , Preescolar , Análisis Mutacional de ADN , Proteínas de Unión al ADN/metabolismo , Femenino , Haplotipos , Humanos , Masculino , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Empalme del ARN , Proteínas Supresoras de Tumor/metabolismo
15.
Mol Pharmacol ; 78(5): 943-51, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20729302

RESUMEN

Methylating agents are first-line therapeutics for gliomas and malignant melanomas. They attack DNA at various sites, and both O(6)-methylguanine and N-methylated base adducts contribute to the killing response. The mechanism of cellular defense against these agents primarily involves O(6)-methylguanine-DNA methyltransferase (MGMT) and base excision repair (BER). Here, we determined whether a key protein involved in DNA double-strand break (DSB) recognition and signaling, nibrin (NBN alias NBS-1), plays a role in the cellular defense against methylating agents. Comparing NBN mutated fibroblasts and lymphoblastoid cells from patients suffering from Nijmegen breakage syndrome, we show that NBN mutants are clearly more sensitive to N-methyl-N'-nitro-N-nitrosoguanidine and temozolomide than the corresponding wild-type cells. Hypersensitivity was due to the induction of both apoptosis and necrosis. The mismatch repair proteins MSH2, MSH6, MLH1, and PMS2 were expressed at a similar level in the cell lines and BER was not affected by NBN mutation. Because MGMT expression abrogated the hypersensitivity of NBN mutated cells, we conclude that O(6)-methylguanine-derived lesions are responsible for triggering the response. Down-regulation of NBN in melanoma cells by small interfering RNA rendered them more sensitive to temozolomide, suggesting that NBN is a novel modulator of temozolomide sensitivity. Because NBN is part of the MRN complex, which recognizes DSBs, the data strongly indicate that MRN is critically involved in DSB processing after O(6)-methylguanine induction. The data provide first evidence that NBN is involved in the cellular defense against O(6)-methylguanine-inducing agents such as temozolomide and identify NBN as a critical target of methylating anticancer drug resistance.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Proteínas de Ciclo Celular/fisiología , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos , Proteínas Nucleares/fisiología , Apoptosis , Caspasa 7/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Transformada , Línea Celular Tumoral , Metilación de ADN , Dacarbazina/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Guanina/análogos & derivados , Guanina/biosíntesis , Homocigoto , Humanos , Melanoma , Metilnitronitrosoguanidina/farmacología , Mutágenos/farmacología , Mutación , Necrosis , Síndrome de Nijmegen/patología , Proteínas Nucleares/genética , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Temozolomida , Neoplasias de la Úvea
16.
Hum Mutat ; 31(9): 1059-68, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20597108

RESUMEN

We have previously shown that mutations in the genes encoding DNA Ligase IV (LIGIV) and RAD50, involved in DNA repair by nonhomologous-end joining (NHEJ) and homologous recombination, respectively, lead to clinical and cellular features similar to those of Nijmegen Breakage Syndrome (NBS). Very recently, a new member of the NHEJ repair pathway, NHEJ1, was discovered, and mutations in patients with features resembling NBS were described. Here we report on five patients from four families of different ethnic origin with the NBS-like phenotype. Sequence analysis of the NHEJ1 gene in a patient of Spanish and in a patient of Turkish origin identified homozygous, previously reported mutations, c.168C>G (p.Arg57Gly) and c.532C>T (p.Arg178Ter), respectively. Two novel, paternally inherited truncating mutations, c.495dupA (p.Asp166ArgfsTer20) and c.526C>T (p.Arg176Ter) and two novel, maternal genomic deletions of 1.9 and 6.9 kb of the NHEJ1 gene, were found in a compound heterozygous state in two siblings of German origin and in one Malaysian patient, respectively. Our findings confirm that patients with NBS-like phenotypes may have mutations in the NHEJ1 gene including multiexon deletions, and show that considerable clinical variability could be observed even within the same family.


Asunto(s)
Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Mutación/genética , Síndrome de Nijmegen/genética , Síndrome de Nijmegen/patología , Secuencia de Bases , Western Blotting , Ciclo Celular , Niño , Preescolar , Inestabilidad Cromosómica/genética , Cromosomas Humanos/genética , Análisis Mutacional de ADN , Genoma Humano/genética , Homocigoto , Humanos , Lactante , Datos de Secuencia Molecular , Fenotipo , Polimorfismo de Nucleótido Simple/genética
17.
Gene ; 447(1): 12-7, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19635536

RESUMEN

Patients affected by the autosomal recessive Nijmegen Breakage Syndrome (NBS [MIM 251260]) have possibly the highest risk for developing a malignancy of all the chromosomal instability syndromes. This reflects the profound disturbance to genomic integrity and cellular homeostasis that is caused by the mutation of the essential mammalian gene, NBN. Whilst null-mutation of Nbn is lethal in the mouse, NBS patients survive due to the fact that the common human founder mutation, found in over 90% of patients, is in fact hypomorphic and leads, by alternative translation, to varying amounts of a partially functional carboxy-terminal protein fragment, p70-nibrin. The expression level of p70-nibrin correlates with cancer incidence amongst patients. Using real-time PCR we have now found that the variation in p70-nibrin expression cannot be attributed to differences in mRNA quantity and that nonsense-mediated mRNA decay is not responsible for the observed variation. We discuss an alternative explanation for p70-nibrin expression variation.


Asunto(s)
Alelos , Proteínas de Ciclo Celular/genética , Síndrome de Nijmegen/genética , Proteínas Nucleares/genética , Proteínas de Ciclo Celular/metabolismo , Reparación del ADN , Predisposición Genética a la Enfermedad , Heterocigoto , Homocigoto , Humanos , Proteínas Nucleares/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética , Células Tumorales Cultivadas
18.
Cell Cycle ; 8(11): 1725-32, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19411856

RESUMEN

The hSNM1B/Apollo protein is involved in the cellular response to DNA-damage as well as in the maintenance of telomeres during S-phase. TRF2 has been shown to interact physically with hSNM1B. As a core component of shelterin, TRF2 functions in organization and protection of telomeres. However, TRF2 was also shown to have a role in the early DNA-damage response, suggesting that hSNM1B and TRF2 cooperate in this dual function. Here we have used Tandem-Affinity-Purification in combination with mass spectrometry to identify additional binding partners of hSNM1B. This revealed HSC70, HSP72, HSP60 and beta-Tubulin to be hSNM1B-interactors. We have confirmed the interaction of hSNM1B and HSP70 in co-immunoprecipitation assays and found that hSNM1B binds to a C-terminal fragment of HSP72, known to contain the substrate binding domain. Depletion of HSP72 in human fibroblasts resulted in a significant reduction of nuclear hSNM1B foci. We also found the phosphorylation of CHK1 at serine 317 to be attenuated in response to UVC irradiation as a consequence of hSNM1B depletion, a result which extends our previous findings on the DNA-damage response function of hSNM1B. HSP70 chaperones have been implicated in the maintenance of genome stability and their expression is often aberrant in cancer. Our results presented here, suggest that the role in genome stability might not be specific to HSP70 but rather can be attributed, at least in part, to hSNM1B. This, together with its stimulating effect on ATM and ATR substrate phosphorylation in response to DNA-damage qualify hSNM1B as a putative target in cancer therapy.


Asunto(s)
Daño del ADN , Enzimas Reparadoras del ADN/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Proteínas de Unión al ADN/metabolismo , Exodesoxirribonucleasas , Proteínas del Choque Térmico HSP72/metabolismo , Humanos , Unión Proteica , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/metabolismo , Fase S , Tubulina (Proteína)/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Rayos Ultravioleta
19.
PLoS One ; 4(5): e5423, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19412544

RESUMEN

BACKGROUND: The NBN gene codes for the protein nibrin, which is involved in the detection and repair of DNA double strand breaks (DSBs). The NBN gene is essential in mammals. METHODOLOGY/PRINCIPAL FINDINGS: We have used a conditional null mutant mouse model in a proteomics approach to identify proteins with modified expression levels after 4 Gy ionizing irradiation in the absence of nibrin in vivo. Altogether, amongst approximately 8,000 resolved proteins, 209 were differentially expressed in homozygous null mutant mice in comparison to control animals. One group of proteins significantly altered in null mutant mice were those involved in oxidative stress and cellular redox homeostasis (p<0.0001). In substantiation of this finding, analysis of Nbn null mutant fibroblasts indicated an increased production of reactive oxygen species following induction of DSBs. CONCLUSIONS/SIGNIFICANCE: In humans, biallelic hypomorphic mutations in NBN lead to Nijmegen breakage syndrome (NBS), an autosomal recessive genetic disease characterised by extreme radiosensitivity coupled with growth retardation, immunoinsufficiency and a very high risk of malignancy. This particularly high cancer risk in NBS may be attributable to the compound effect of a DSB repair defect and oxidative stress.


Asunto(s)
Reparación del ADN , Proteínas Nucleares/deficiencia , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/genética , Reparación del ADN/fisiología , Proteínas de Unión al ADN , Electroforesis en Gel Bidimensional , Expresión Génica , Respuesta al Choque Térmico , Homeostasis , Hígado/metabolismo , Hígado/efectos de la radiación , Ratones , Ratones Noqueados , Chaperonas Moleculares/metabolismo , NAD/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Proteoma/aislamiento & purificación , Proteoma/metabolismo , Proteómica , Especies Reactivas de Oxígeno/metabolismo
20.
DNA Repair (Amst) ; 8(3): 424-8, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19073284

RESUMEN

The Tenth Meeting of the German Society for Research on DNA Repair was held in Berlin in September 2008. Invited presentations by Yosef Shiloh, Stanton L. Gerson, Sacha Beneke, Patrick Concannon, Jochen Dahm-Daphi, Thilo Dörk, Friedrike Eckardt-Schupp, Bernd Epe, Ian Hickson, Ulrich Hübscher, Penny Jeggo, Malik Lutzmann, Christof Niehrs, Primo Schär and Predrag Slijepcevic together with over 80 selected oral and poster presentations generated an inspiring scientific program, which documented the impressive progress of the community and defined future challenges in the field.


Asunto(s)
Reparación del ADN , Alemania
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...