Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Genet Med ; 20(4): 458-463, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28837157

RESUMEN

PurposeMutations in genes involved in Fanconi anemia (FA)/BRCA DNA repair pathway cause cancer susceptibility diseases including familial breast cancer and Fanconi anemia (FA). A single FA patient with biallelic FANCM mutations was reported in 2005 but concurrent FANCA pathogenic mutations precluded assignment of FANCM as an FA gene. Here we report three individuals with biallelic FANCM truncating mutations who developed early-onset cancer and toxicity to chemotherapy but did not present congenital malformations or any hematological phenotype suggestive of FA.MethodsChromosomal breakages, interstrand crosslink sensitivity, and FANCD2 monoubiquitination were assessed in primary fibroblasts. Mutation analysis was achieved through Sanger sequencing. Genetic complementation of patient-derived cells was performed by lentiviral mediated transduction of wild-type FANCM complementary DNA followed by functional studies.ResultsPatient-derived cells exhibited chromosomal fragility, hypersensitivity to interstrand crosslinks, and impaired FANCD2 monoubiquitination. We identified two homozygous mutations (c.2586_2589del4; p.Lys863Ilefs*12 and c.1506_1507insTA; p.Ile503*) in FANCM as the cause of the cellular phenotype. Patient-derived cells were genetically complemented upon wild-type FANCM complementary DNA expression.ConclusionLoss-of-function mutations in FANCM cause a cancer predisposition syndrome clinically distinct from bona fide FA. Care should be taken with chemotherapy and radiation treatments in these patients due to expected acute toxicity.


Asunto(s)
Alelos , ADN Helicasas/genética , Anemia de Fanconi/genética , Predisposición Genética a la Enfermedad , Neoplasias/diagnóstico , Neoplasias/genética , Eliminación de Secuencia , Adolescente , Línea Celular , Fragilidad Cromosómica/efectos de los fármacos , ADN Helicasas/metabolismo , Femenino , Estudios de Asociación Genética , Prueba de Complementación Genética , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Linaje , Fenotipo
2.
Genome Res ; 25(3): 402-12, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25609572

RESUMEN

We have previously demonstrated that in Saccharomyces cerevisiae replication, checkpoint inactivation via a mec1 mutation leads to chromosome breakage at replication forks initiated from virtually all origins after transient exposure to hydroxyurea (HU), an inhibitor of ribonucleotide reductase. Here we sought to determine whether all replication forks containing single-stranded DNA gaps have equal probability of producing double-strand breaks (DSBs) when cells attempt to recover from HU exposure. We devised a new methodology, Break-seq, that combines our previously described DSB labeling with next generation sequencing to map chromosome breaks with improved sensitivity and resolution. We show that DSBs preferentially occur at genes transcriptionally induced by HU. Notably, different subsets of the HU-induced genes produced DSBs in MEC1 and mec1 cells as replication forks traversed a greater distance in MEC1 cells than in mec1 cells during recovery from HU. Specifically, while MEC1 cells exhibited chromosome breakage at stress-response transcription factors, mec1 cells predominantly suffered chromosome breakage at transporter genes, many of which are the substrates of those transcription factors. We propose that HU-induced chromosome fragility arises at higher frequency near HU-induced genes as a result of destabilized replication forks encountering transcription factor binding and/or the act of transcription. We further propose that replication inhibitors can induce unscheduled encounters between replication and transcription and give rise to distinct patterns of chromosome fragile sites.


Asunto(s)
Fragilidad Cromosómica/efectos de los fármacos , Roturas del ADN de Doble Cadena , Replicación del ADN , Secuenciación de Nucleótidos de Alto Rendimiento , Hidroxiurea/farmacología , Transcripción Genética , Ciclo Celular/genética , Rotura Cromosómica , Regulación Fúngica de la Expresión Génica , Genes Fúngicos , Iones/metabolismo , Proteínas de Transporte de Membrana/genética , Metales/metabolismo , Origen de Réplica , Estrés Fisiológico , Factores de Transcripción/metabolismo , Levaduras/efectos de los fármacos , Levaduras/genética , Levaduras/metabolismo
3.
BMC Med Genomics ; 7: 24, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24885126

RESUMEN

BACKGROUND: Fanconi anemia (FA) is a rare inherited genetic syndrome with highly variable clinical manifestations. Fifteen genetic subtypes of FA have been identified. Traditional complementation tests for grouping studies have been used generally in FA patients and in stepwise methods to identify the FA type, which can result in incomplete genetic information from FA patients. METHODS: We diagnosed five pediatric patients with FA based on clinical manifestations, and we performed exome sequencing of peripheral blood specimens from these patients and their family members. The related sequencing data were then analyzed by bioinformatics, and the FANC gene mutations identified by exome sequencing were confirmed by PCR re-sequencing. RESULTS: Homozygous and compound heterozygous mutations of FANC genes were identified in all of the patients. The FA subtypes of the patients included FANCA, FANCM and FANCD2. Interestingly, four FA patients harbored multiple mutations in at least two FA genes, and some of these mutations have not been previously reported. These patients' clinical manifestations were vastly different from each other, as were their treatment responses to androstanazol and prednisone. This finding suggests that heterozygous mutation(s) in FA genes could also have diverse biological and/or pathophysiological effects on FA patients or FA gene carriers. Interestingly, we were not able to identify de novo mutations in the genes implicated in DNA repair pathways when the sequencing data of patients were compared with those of their parents. CONCLUSIONS: Our results indicate that Chinese FA patients and carriers might have higher and more complex mutation rates in FANC genes than have been conventionally recognized. Testing of the fifteen FANC genes in FA patients and their family members should be a regular clinical practice to determine the optimal care for the individual patient, to counsel the family and to obtain a better understanding of FA pathophysiology.


Asunto(s)
Análisis Mutacional de ADN/métodos , Exoma/genética , Anemia de Fanconi/genética , Mutación/genética , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Niño , Preescolar , Fragilidad Cromosómica/efectos de los fármacos , Fragilidad Cromosómica/genética , Ensayo Cometa , Anemia de Fanconi/diagnóstico , Femenino , Humanos , Cariotipificación , Masculino , Mitomicina/farmacología , Reproducibilidad de los Resultados , Programas Informáticos , Estadística como Asunto
4.
J Cell Sci ; 126(Pt 11): 2502-15, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23549790

RESUMEN

Transcription factor IIH (TFIIH) participates in transcription, nucleotide excision repair and the control of the cell cycle. In the present study, we demonstrate that the Dmp52 subunit of TFIIH in Drosophila physically interacts with the fly p53 homologue, Dp53. The depletion of Dmp52 in the wing disc generates chromosome fragility, increases apoptosis and produces wings with a reduced number of cells; cellular proliferation, however, is not affected. Interestingly, instead of suppressing the apoptotic phenotype, the depletion of Dp53 in Dmp52-depleted wing disc cells increases apoptosis and the number of cells that suffer from chromosome fragility. The apoptosis induced by the depletion of Dmp52 alone is partially dependent on the JNK pathway. In contrast, the enhanced apoptosis caused by the simultaneous depletion of Dp53 and Dmp52 is absolutely JNK-dependent. In this study, we also show that the anti-proliferative drug triptolide, which inhibits the ATPase activity of the XPB subunit of TFIIH, phenocopies the JNK-dependent massive apoptotic phenotype of Dp53-depleted wing disc cells; this observation suggests that the mechanism by which triptolide induces apoptosis in p53-deficient cancer cells involves the activation of the JNK death pathway.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Diterpenos/farmacología , Proteínas de Drosophila/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Fenantrenos/farmacología , Factor de Transcripción TFIIH/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/genética , Fragilidad Cromosómica/efectos de los fármacos , Fragilidad Cromosómica/genética , Cromosomas de Insectos/genética , Cromosomas de Insectos/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster , Compuestos Epoxi/farmacología , MAP Quinasa Quinasa 4/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Factor de Transcripción TFIIH/genética , Proteína p53 Supresora de Tumor/genética
5.
C R Biol ; 336(1): 29-33, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23537767

RESUMEN

Fanconi anemia (FA) is a recessive chromosomal instability syndrome that is clinically characterized by multiple symptoms. Chromosome breakage hypersensitivity to alkylating agents is the gold standard test for FA diagnosis. In this study, we provide a detailed laboratory protocol for accurate assessment of FA diagnosis based on mitomycin C (MMC) test. Induced chromosomal breakage study was successful in 171 out of 205 aplastic anemia (AA) patients. According to the sensitivity of MMC at 50 ng/ml, 38 patients (22.22%) were diagnosed as affected and 132 patients (77.17%) as unaffected. Somatic mosaicism was suspected in an 11-year-old patient with a FA phenotype. Twenty-six siblings of FA patients were also evaluated and five of them (19.23%) were diagnosed as FA. From this study, a standard protocol for diagnosis of FA was developed. It is routinely used as a diagnostic test of FA in Tunisia.


Asunto(s)
Anemia Aplásica/diagnóstico , Antibióticos Antineoplásicos , Anemia de Fanconi/diagnóstico , Mitomicina , Adolescente , Adulto , Anemia Aplásica/epidemiología , Anemia Aplásica/genética , Niño , Preescolar , Rotura Cromosómica/efectos de los fármacos , Fragilidad Cromosómica/efectos de los fármacos , Consanguinidad , Diagnóstico Diferencial , Anemia de Fanconi/epidemiología , Anemia de Fanconi/genética , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mosaicismo , Túnez/epidemiología , Adulto Joven
6.
J Appl Genet ; 53(2): 221-6, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22415351

RESUMEN

Fifty river buffalo (Bubalus bubalis, 2n = 50) cows reared in two different provinces of Campania (southern Italy) underwent cytogenetic investigations to ascertain possible differences in their chromosome stability. One group (Caserta province) was under legal sequestration due to the presence in the milk mass of higher mean values of dioxins [21.79 pg/g of fat as sum of polychloro-dibenzo-dioxins (PCDDs), polychloro-dibenzo-furans (PCDFs) and dioxin-like polychlorobiphenyls (DL-PCBs)] than both those permitted (6.0 pg/g of fat as WHO-TEQ) and those (1.3 pg/g of fat as WHO-TEQ) observed in the control group raised in Salerno province. Two types of peripheral blood cell cultures were performed: without (normal cultures for the chromosome abnormality (CA) test: chromatid breaks, chromosome breaks, fragments) and with the addition of BrdU for the sister chromatid exchange (SCE) test). The CA test revealed a significantly (P < 0.01) higher chromosome fragility in the exposed cows compared to the control. Indeed, mean values of CA/cell were 1.26 ± 1.15 in exposed cows and 0.37 ± 0.71 in the control. Mean SCE was higher in exposed cows (8.50 ± 3.35) than that (8.29 ± 3.51) found in the control but the difference was not significant. Comparison within the same group of cows at first (FL) and multiple (ML) lactations revealed significantly (P < 0.01) higher mean values of CA/cell in exposed ML-cows vs FL-cows while no statistical differences were found between ML-cows and FL-cows in the control farm. By contrast, significantly (P < 0.01) higher mean values of SCE were found in both groups of FL-cows versus ML-cows. Comparisons with other previous studied species (sheep and cattle) were also performed.


Asunto(s)
Búfalos/genética , Aberraciones Cromosómicas/efectos de los fármacos , Fragilidad Cromosómica/efectos de los fármacos , Dioxinas/análisis , Contaminantes Ambientales/análisis , Leche/química , Animales , Bromodesoxiuridina , Bovinos , Células Cultivadas , Fragilidad Cromosómica/genética , Dioxinas/toxicidad , Contaminantes Ambientales/toxicidad , Femenino , Italia , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Oveja Doméstica/genética , Intercambio de Cromátides Hermanas/efectos de los fármacos , Intercambio de Cromátides Hermanas/genética
7.
Mutagenesis ; 26(2): 269-72, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20952519

RESUMEN

In this study, we compared cross-bred dairy cows in the Susa Valley (Piedmont, northern Italy), reared either near a high-temperature steel production plant (Farms A and B) or in an industry-free area (control). Exposed cows (n = 36) were selected based on mean bulk milk toxic equivalent values of polychlorodibenzodioxins (PCDDs) and dioxin-like (DL) polychlorobiphenyls (PCBs) and polychlorodibenzofurans (PCDFs) equal to 18.56 pg/g fat and 8.56 pg/g of fat in dairy cows from Farms A and B, respectively, exceeding both those permitted by the legislation in force (6 pg/g fat PCDDs and DL-PCDFs/PCBs), and those measured in dairy cows (n = 19) of the farm used as control (1.75 pg/g of fat PCDDs and DL-PCDFs/PCBs). Two types of peripheral blood cell cultures were performed: without (normal cultures for the chromosome abnormality (CA)-test: gaps, chromatid breaks, chromosome breaks and fragments) and with addition of bromodeoxyuridine [for the sister chromatid exchange (SCE)-test]. Both tests revealed a significant (P ≤ 0.05) higher chromosome fragility in the exposed cattle compared to controls: CA/cell mean values (without gaps) were 0.65 ± 0.91, 0.51 ± 0.81 and 0.13 ± 0.39 in Farms A, B and controls, respectively, while SCE/cell mean values were 7.00 ± 2.88, 6.39 ± 2.80 and 5.29 ± 2.51. Although the role of other pollutants (e.g. heavy metals) in the genesis of the recorded chromosome alterations cannot be ruled out, our results confirm the findings of previous research into dioxin-exposed sheep.


Asunto(s)
Benzofuranos/toxicidad , Fragilidad Cromosómica/efectos de los fármacos , Dioxinas/toxicidad , Contaminantes Ambientales/toxicidad , Mutágenos/toxicidad , Bifenilos Policlorados/toxicidad , Polímeros/toxicidad , Animales , Bovinos , Células Cultivadas , Rotura Cromosómica/efectos de los fármacos , Cariotipificación , Leche/química , Intercambio de Cromátides Hermanas/efectos de los fármacos
8.
Chromosoma ; 119(6): 575-87, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20585795

RESUMEN

The replication dynamics at common fragile site FRA6E has been evaluated by molecular combing and interphase fluorescent in situ hybridisation (FISH) in primary human lymphocytes cultured under normal or aphidicolin-induced stress conditions. FRA6E is one of the most frequently expressed common fragile sites of the human genome. It harbours several genes, PARK2 being regarded as the most relevant one. According to the results obtained from interphase FISH analysis, FRA6E can be considered a mid-late-replicating sequence characterised by heterogeneous replication timing. Molecular combing did not reveal specific replication parameters at the fragile site: fork rates were highly comparable to those detected at an early replicating locus (LMNB2) used as control and in very good agreement with the whole-genome data obtained in parallel. The same indication applied to the density of initiation zones, the inter-origin distances from adjacent ongoing forks, the frequencies of unidirectional forks, fork arrest events and asynchronous forks. Interestingly, PARK2 appeared embedded in an early/late replication transition zone, corresponding to intron 8 (162 kb) and to the fragility core of FRA6E. In cells exposed to aphidicolin, few forks progressing at a rather slow rate were observed, the majority of them being unidirectional, but again a specific response of the fragile site was not observed. In summary, at FRA6E the replication process is not impaired per se, but chromosome breakages occur preferentially at an early/late replication transition zone. Aphidicolin might increase the occurrence of breakage events at FRA6E by prolonging the time interval separating the replication of early and late replication domains. These results may be of general significance to address the problem of fragile site instability.


Asunto(s)
Sitios Frágiles del Cromosoma , Replicación del ADN , Afidicolina/farmacología , Ciclo Celular , Células Cultivadas , Rotura Cromosómica , Sitios Frágiles del Cromosoma/efectos de los fármacos , Fragilidad Cromosómica/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Humanos , Hibridación Fluorescente in Situ , Linfocitos/citología , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
9.
Mutat Res ; 695(1-2): 46-54, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19932191

RESUMEN

The antihypertensive drug atenolol was found to induce chromosome loss, detected as micronuclei in the peripheral lymphocytes of treated patients. The fundamental question which chromosomes the micronuclei were derived from remains to be answered. Analysis of structural chromosomal aberrations (CAs) and expression of fragile sites (FS) were pursued in this study. They revealed a significantly higher incidence of chromosomal aberrations (chromatid and chromosome breaks) in patients compared with controls, where 10 FS emerged as specific. Also, the band 17q12-21, where known fragile sites have not been reported, was only expressed in atenolol-treated patients. Fluorescence in situ hybridization using chromosome-specific probes revealed the preferential involvement of chromosomes 7, 11, 17 and X in the micronuclei (MN) of patients. The results also suggest a correlation between chromosomal fragility and content of MN, and support the findings for a linkage between hypertension and a locus on chromosome 17.


Asunto(s)
Antihipertensivos/toxicidad , Atenolol/toxicidad , Fragilidad Cromosómica/efectos de los fármacos , Daño del ADN , Hipertensión/tratamiento farmacológico , Linfocitos/efectos de los fármacos , Micronúcleos con Defecto Cromosómico/efectos de los fármacos , Adulto , Anciano , Estudios de Casos y Controles , Sitios Frágiles del Cromosoma , Cromosomas Humanos Par 17/genética , Daño del ADN/efectos de los fármacos , Femenino , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Masculino , Pruebas de Micronúcleos , Persona de Mediana Edad
10.
Yonsei Med J ; 50(1): 137-41, 2009 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-19259360

RESUMEN

PURPOSE: Genetic factors are known to be important in the etiology of bipolar disorder (BD). The fragile sites (FSs) are a very interesting subject for the study of clinical disorders. The aim of this study was to evaluate fragile sites seen in patients with bipolar disorder and find a correlation between some fragile sites and bipolar disorder. PATIENTS AND METHODS: The frequencies of folate sensitive FSs were compared in short-term whole blood cultures from bipolar patients and from normal individuals. RESULTS: The rate of FS expression in the patients was considerably higher than in the controls (p < 0.001). Several chromosome regions including 1p36, 1q21, 1q32, 3p25, 7q22, 7q32, 11q23, 12q24, 13q32, 14q24, Xp22 and Xq26 were represented considerably more often in the patients than in the controls (p value between 0.001 to 0.036). Among these FSs, the sites 1p36, 1q21, 3p25, 7q22, 7q32, and 14q24 were not observed in other studies. CONCLUSION: These regions can be the most active of hot spots in the genomes of bipolar patients, and may harbor important genes associated with BD.


Asunto(s)
Trastorno Bipolar/genética , Sitios Frágiles del Cromosoma/genética , Fragilidad Cromosómica/genética , Cromosomas Humanos/genética , Adolescente , Adulto , Sitios Frágiles del Cromosoma/efectos de los fármacos , Fragilidad Cromosómica/efectos de los fármacos , Citogenética , Femenino , Ácido Fólico/farmacología , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
11.
Yonsei Medical Journal ; : 137-141, 2009.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-52281

RESUMEN

PURPOSE: Genetic factors are known to be important in the etiology of bipolar disorder (BD). The fragile sites (FSs) are a very interesting subject for the study of clinical disorders. The aim of this study was to evaluate fragile sites seen in patients with bipolar disorder and find a correlation between some fragile sites and bipolar disorder. PATIENTS AND METHODS: The frequencies of folate sensitive FSs were compared in short-term whole blood cultures from bipolar patients and from normal individuals. RESULTS: The rate of FS expression in the patients was considerably higher than in the controls (p < 0.001). Several chromosome regions including 1p36, 1q21, 1q32, 3p25, 7q22, 7q32, 11q23, 12q24, 13q32, 14q24, Xp22 and Xq26 were represented considerably more often in the patients than in the controls (p value between 0.001 to 0.036). Among these FSs, the sites 1p36, 1q21, 3p25, 7q22, 7q32, and 14q24 were not observed in other studies. CONCLUSION: These regions can be the most active of hot spots in the genomes of bipolar patients, and may harbor important genes associated with BD.


Asunto(s)
Adolescente , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Trastorno Bipolar/genética , Sitios Frágiles del Cromosoma/efectos de los fármacos , Fragilidad Cromosómica/efectos de los fármacos , Cromosomas Humanos/genética , Citogenética , Ácido Fólico/farmacología , Predisposición Genética a la Enfermedad
12.
Biochim Biophys Acta ; 1779(1): 3-16, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18078840

RESUMEN

Fragile sites are heritable specific chromosome loci that exhibit an increased frequency of gaps, poor staining, constrictions or breaks when chromosomes are exposed to partial DNA replication inhibition. They constitute areas of chromatin that fail to compact during mitosis. They are classified as rare or common depending on their frequency within the population and are further subdivided on the basis of their specific induction chemistry into different groups differentiated as folate sensitive or non-folate sensitive rare fragile sites, and as aphidicolin, bromodeoxyuridine (BrdU) or 5-azacytidine inducible common fragile sites. Most of the known inducers of fragility share in common their potentiality to inhibit the elongation of DNA replication, particularly at fragile site loci. Seven folate sensitive (FRA10A, FRA11B, FRA12A, FRA16A, FRAXA, FRAXE and FRAXF) and two non-folate sensitive (FRA10B and FRA16B) fragile sites have been molecularly characterized. All have been found to represent expanded DNA repeat sequences resulting from a dynamic mutation involving the normally occurring polymorphic CCG/CGG trinucleotide repeats at the folate sensitive and AT-rich minisatellite repeats at the non-folate sensitive fragile sites. These expanded repeats were demonstrated, first, to have the potential, under certain conditions, to form stable secondary non-B DNA structures (intra-strand hairpins, slipped strand DNA or tetrahelical structures) and to present highly flexible repeat sequences, both conditions which are expected to affect the replication dynamics, and second, to decrease the efficiency of nucleosome assembly, resulting in decondensation defects seen as fragile sites. Thirteen aphidicolin inducible common fragile sites (FRA2G, FRA3B, FRA4F, FRA6E, FRA6F, FRA7E, FRA7G, FRA7H, FRA7I, FRA8C, FRA9E, FRA16D and FRAXB) have been characterized at a molecular level and found to represent relatively AT-rich DNA areas, but without any expanded repeat motifs. Analysis of structural characteristics of the DNA at some of these sites (FRA2G, FRA3B, FRA6F, FRA7E, FRA7G, FRA7H, FRA7I, FRA16D and FRAXB) showed that they contained more areas of high DNA torsional flexibility with more highly AT-dinucleotide-rich islands than neighbouring non-fragile regions. These islands were shown to have the potential to form secondary non-B DNA structures and to interfere with higher-order chromatin folding. Therefore, a common fragility mechanism, characterized by high flexibility and the potential to form secondary structures and interfere with nucleosome assembly, is shared by all the cloned classes of fragile sites. From the clinical point of view, the folate sensitive rare fragile site FRAXA is the most important fragile site as it is associated with the fragile X syndrome, the most common form of familial mental retardation, affecting about 1/4000 males and 1/6000 females. Mental retardation in this syndrome is considered as resulting from the abolition of the FMR1 gene expression due to hypermethylation of the gene CpG islands adjacent to the expanded methylated trinucleotide repeat. FRAXE is associated with X-linked non-specific mental retardation, and FRA11B with Jacobsen syndrome. There is also some evidence that fragile sites, especially common fragile sites, are consistently involved in the in vivo chromosomal rearrangements related to cancer, whereas the possible implication of common fragile sites in neuropsychiatric and developmental disorders is still poorly documented.


Asunto(s)
Sitios Frágiles del Cromosoma , Fragilidad Cromosómica , Secuencia de Bases , Sitios Frágiles del Cromosoma/efectos de los fármacos , Fragilidad Cromosómica/efectos de los fármacos , Citogenética , ADN/genética , Distamicinas/farmacología , Femenino , Ácido Fólico/farmacología , Síndrome del Cromosoma X Frágil/genética , Expresión Génica , Humanos , Masculino , Expansión de Repetición de Trinucleótido
13.
Int J Hematol ; 85(4): 354-61, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17483082

RESUMEN

Designing stem cell transplantation (SCT) conditioning regimens for Fanconi anemia (FA) has proved difficult because of hypersensitivity to the DNA cross-linking agents. We performed chromosome fragility tests with 56 FA patients and with 50 non-FA patients with severe aplastic anemia or myelodysplastic syndrome. We evaluated peripheral blood lymphocyte specimens cultured for 72 hours and treated with mitomycin C, diepoxybutane (DEB), cyclophosphamide (CY) metabolites, cytosine arabinoside (Ara-C), and fludarabine (Flu) metabolite (9-beta-D-arabinofuranosyl-2-fluoroadenine [2-F-Ara-A]). The DEB and CY metabolite tests were highly sensitive and specific for FA (P<10(-4)) for both tests), and the number of aberrations per cell for DEB correlated with that for the CY metabolite test (P < 10(-4)) but did not correlate with the number of aberrations per cell for the Ara-C and 2-F-Ara-A tests. The difference in breakage frequencies between FA and non-FA patients for cultures treated with 2-F-Ara-A was not statistically significant. Most of the breakages observed in cells treated with 2-F-Ara-A-and Ara-C were chromatid breaks. It may be possible to determine the appropriate CY dose in the preconditioning regimen for SCT in FA patients on the basis of the in vitro effects on fragility, and Flu or Ara-C may be a safer drug than high-dose CY for conditioning in FA patients.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Rotura Cromosómica/efectos de los fármacos , Ciclofosfamida/farmacología , Citarabina/farmacología , Anemia de Fanconi/genética , Agonistas Mieloablativos/farmacología , Acondicionamiento Pretrasplante , Vidarabina/análogos & derivados , Células Cultivadas , Fragilidad Cromosómica/efectos de los fármacos , Anemia de Fanconi/metabolismo , Anemia de Fanconi/terapia , Femenino , Humanos , Masculino , Trasplante de Células Madre , Vidarabina/farmacología
14.
Prenat Diagn ; 25(4): 318-21, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15849796

RESUMEN

OBJECTIVE: To report fragility at 10q23.3 in a fetus exposed to phenytoin during pregnancy. Review of the literature. METHODS: Amniocytes were cultured in A10 (WISENT) culture medium. Molecular polymorphism studies of MTHFR gene using PCR were performed on fetal tissues. RESULTS: The fragile site was expressed in all 22 amniocyte colonies analyzed. Analysis of fetal blood showed 46,XX[98]/46,XX,fra(10)(q23.3)[3]/46,XX,del(10)(q23.3) [1]. Molecular studies of the MTHFR (methylenetetrahydrofolate reductase) gene identified a compound heterozygote genotype for two polymorphisms, 677C>T and 1298A>C. CONCLUSION: The fragility at 10q23.3 is unlikely to be due to culture condition-induced folic acid deficiency (medium contains folate). It is possible that this finding represents a previously undescribed folic acid-insensitive fragile site in the region of 10q23.3. Alternatively, the fetal cells may have had decreased folate metabolism, and the fragile site was the known folate-sensitive FRA10A. Since phenytoin has been shown to decrease MTHFR activity in mice, we postulate that the fragile site at 10q23.3 in this fetus may have arisen secondary to a combination of the polymorphisms in MTHFR and exposure to this drug, and is indeed FRA10A.


Asunto(s)
Amniocentesis , Anticonvulsivantes/uso terapéutico , Sitios Frágiles del Cromosoma/genética , Fragilidad Cromosómica/genética , Cromosomas Humanos Par 10 , Fenitoína/uso terapéutico , Adulto , Células Cultivadas , Deleción Cromosómica , Sitios Frágiles del Cromosoma/efectos de los fármacos , Fragilidad Cromosómica/efectos de los fármacos , Epilepsia/tratamiento farmacológico , Femenino , Humanos , Cariotipificación , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Reacción en Cadena de la Polimerasa , Polimorfismo Genético , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico
15.
Aquat Toxicol ; 66(3): 319-29, 2004 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-15129773

RESUMEN

This review summarises the wide range of toxicological effects of malachite green (MG), a triarylmethane dye on various fish species and certain mammals. MG is widely used in aquaculture as a parasiticide and in food, health, textile and other industries for one or the other purposes. It controls fungal attacks, protozoan infections and some other diseases caused by helminths on a wide variety of fish and other aquatic organisms. However, the dye has generated much concern regarding its use, due to its reported toxic effects. The toxicity of this dye increases with exposure time, temperature and concentration. It has been reported to cause carcinogenesis, mutagenesis, chromosomal fractures, teratogenecity and respiratory toxicity. Histopathological effects of MG include multi-organ tissue injury. Significant alterations occur in biochemical parameters of blood in MG exposed fish. Residues of MG and its reduced form, leucomalachite green have been reported from serum, liver, kidney, muscles and other tissues as also from eggs and fry. Toxicity occurs in some mammals, including organ damage, mutagenic, carcinogenic and developmental abnormalities. However, despite the large amount of data on its toxic effects, MG is still used as a parasiticide in aquaculture and other industries. It is concluded that the potential of alternative parasiticides, like humic acid, chlorine dioxide and Pyceze, should be explored to replace MG. Until then, MG should be used with extreme care at suitable concentrations and at times when the temperature is low. Removal of residual MG in treatment ponds should also be considered.


Asunto(s)
Carcinógenos/toxicidad , Fragilidad Cromosómica/efectos de los fármacos , Colorantes/toxicidad , Peces/metabolismo , Fungicidas Industriales/toxicidad , Mutagénesis/efectos de los fármacos , Colorantes de Rosanilina/toxicidad , Compuestos de Anilina , Animales , Acuicultura/métodos , Dosificación Letal Mediana , Temperatura , Factores de Tiempo
16.
Nucleic Acids Res ; 31(23): 6733-40, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14627806

RESUMEN

The molecular basis of the myotonic dystrophy type 1 is the expansion of a CTG repeat at the DMPK locus. The expanded disease-associated repeats are unstable in both somatic and germ lines, with a high tendency towards expansion. The rate of expansion is directly related to the size of the pathogenic allele, increasing the size heterogeneity with age. It has also been suggested that additional factors, including as yet unidentified environmental factors, might affect the instability of the expanded CTG repeats to account for the observed CTG size dynamics over time. To investigate the effect of environmental factors in the CTG repeat instability, three lymphoblastoid cell lines were established from two myotonic dystrophy patients and one healthy individual, and parallel cultures were concurrently expanded in the presence or absence of the mutagenic chemical mitomycin C for a total of 12 population doublings. The new alleles arising along the passages were analysed by radioactive small pool PCR and sequencing gels. An expansion bias of the stepwise mutation was observed in a (CTG)124 allele of a cell line harbouring two modal alleles of 28 and 124 CTG repeats. Interestingly, this expansion bias was clearly enhanced in the presence of mitomycin C. The effect of mitomycin C was also evident in the normal size alleles in two cell lines with alleles of 13/13 and 12/69 repeats, where treated cultures showed new longer alleles. In conclusion, our results indicate that mitomycin C modulates the dynamics of myotonic dystrophy-associated CTG repeats in LBCLs, enhancing the expansion bias of long-pathogenic repeats and promoting the expansion of normal length repeats.


Asunto(s)
Fragilidad Cromosómica/efectos de los fármacos , Mitomicina/farmacología , Mutagénesis/efectos de los fármacos , Distrofia Miotónica/genética , Expansión de Repetición de Trinucleótido/efectos de los fármacos , Alelos , Células Cultivadas , Fragilidad Cromosómica/genética , Humanos , Expansión de Repetición de Trinucleótido/genética
17.
Chromosoma ; 97(6): 459-64, 1989 May.
Artículo en Inglés | MEDLINE | ID: mdl-2501072

RESUMEN

Nineteen fragile sites induced by aphidicolin in lymphocyte cultures from the laboratory mouse are documented. These sites are compared with previously described fragile sites induced in mouse fibroblast systems, and then with those reported on chromosomes which have been evolutionarily conserved between the mouse and the laboratory rat. Of a total of 38 fragile sites thus far identified in mouse fibroblasts and lymphocytes, only 4 sites are common to the two cell types; 34 sites show no correspondence of loci. The reason for this discrepancy is unclear, but it is possible that these data may indicate some degree of tissue specificity of fragile site expression in the mouse. Eight autosomes in the mouse and rat retain straightforward and nearly complete banding homology. To test the hypothesis that fragile sites are conserved between the two species, we compared these eight autosomes with regard to number and distribution of fragile site loci. A total of 30 fragile sites is distributed over the conserved chromosomes. Only 4 (possibly 5) are common to both species; 18 are found in the rat but not the mouse, and 4 are found in the mouse but not the rat. Of the 4 shared sites, notable differences in frequency of expression exist. Our comparisons show that: (1) a small number of fragile sites is conserved; (2) a large number of fragile sites is not conserved, and (3) some sites which are conserved are quite different in the frequency at which they are expressed in the two species, indicating that the sites themselves may have undergone evolutionary change.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Fragilidad Cromosómica , Ratones Endogámicos ICR/genética , Ratas/genética , Animales , Afidicolina , Evolución Biológica , Sitios Frágiles del Cromosoma , Fragilidad Cromosómica/efectos de los fármacos , Diterpenos/farmacología , Femenino , Masculino , Ratones , Especificidad de la Especie
18.
Am J Med Genet ; 32(4): 514-9, 1989 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2505618

RESUMEN

Analysis of chromosome breakage with mitomycin C (MMC) and folate-deficient culture conditions was undertaken on 18 Prader-Labhart-Willi syndrome (PLWS) patients (10 with 15q12 deletion [5 females, 5 males; mean age = 17.9 yr, range of 0.3 to 40 yr] and 8 without deletion [2 females, 6 males; mean age = 18.6 yr, range of 7 to 26 yr]), 21 PLWS parents with an average age of 39.2 yr and a range of 25 to 70 yr (12 fathers [8 fathers of PLWS children with the 15q12 deletion and 4 fathers of PLWS children with normal chromosomes] and 9 mothers [4 mothers of PLWS children with the 15q12 deletion and 5 mothers of PLWS children with normal chromosomes]), and age-matched control individuals. There was no difference between PLWS patients and control individuals in the number of chromosome and chromatid aberrations in cells grown at 48 and/or 96 hr in either 20 ng/ml or 50 ng/ml of MMC or between the PLWS parents and control individuals in cells grown in 50 ng/ml MMC for 96 hr, although a small increase (P less than 0.05) in chromosome breakage was found in cells from the total PLWS parental group compared with control individuals exposed for 48 hr in 50 ng/ml MMC. There was also no significant difference in chromosome fragile site frequency in cells grown in folate-deficient culture conditions in PLWS patients, their parents, or controls.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Fragilidad Cromosómica , Cromosomas Humanos Par 15 , Síndrome de Prader-Willi/genética , Adolescente , Adulto , Células Cultivadas , Niño , Preescolar , Deleción Cromosómica , Sitios Frágiles del Cromosoma , Fragilidad Cromosómica/efectos de los fármacos , Cromosomas Humanos Par 15/efectos de los fármacos , Femenino , Ácido Fólico/farmacología , Pruebas Genéticas , Humanos , Lactante , Masculino , Mitomicina , Mitomicinas/farmacología , Intercambio de Cromátides Hermanas/efectos de los fármacos
19.
Acta bioquím. clín. latinoam ; 21(4): 461-6, dic. 1987. tab
Artículo en Español | LILACS | ID: lil-63892

RESUMEN

En la actualidad se considera que se llega a un estado de malignidad a partir de una secuencia de pasos que se inician con la aparici[on de una c[elulas premaligna (portadora de una mutaci[on), la que desde ese momento se detiene en su estadio de diferenciaci[on, manteniendo su capacidad de divisi[on. La aparici[on posterior de cambios genéticos específicos sería responsable de la transición de células francamente malignas que poseen anomalías cromosómicas características. La obsrevación de anomalías cromosómicas específicas en células neoplásicas corrobora el papel fundamental que tienen estas anomalías en la transformación de una célula normal a otra maligna. Si bien no se conocen con exactitud los mecanismos por los cuales se originan las alteraciones cromosómicas, actualmente se acepta que pueden verse facilitadas por la presencia, entre otros, de los llamados sitios frágiles (SF), observables en los cromosomas (1). La primera mención de SF fue publicada en 1965 por Dekaban, describiendo un clon de células con un cromosoma del grupo C anorormal (2). Tres años más tarde, Lejeune, demostró la heredabilidad de estos sitios (3). En 1969, Lubs citó un SF en el cromosoma X (fra(X)) asociado al retardo mental (4). En los últimos tres años, se han llegado a identificar alrededor de 90 SF, de los cuales solamente el fra(X) está relacionado con una anormalidad fenotípica, mientras que se postula que los restnates pueden actuar como factores predisponentes a rupturas y a rearreglos cromosómicos, muchos de ellos, involucrados en diferentes neoplasias humanas (1). Esta idea se ve apoyada por el hecho de que los puntos de ruptura de ciertos reordenamientos estructurales coinciden con la ubicación cromosómica de determinados sitios frágiles


Asunto(s)
Humanos , Fragilidad Cromosómica , Neoplasias/genética , Aberraciones Cromosómicas , Fragilidad Cromosómica/efectos de los fármacos , Oncogenes
20.
Acta bioquím. clín. latinoam ; 21(4): 461-6, dic. 1987. Tab
Artículo en Español | BINACIS | ID: bin-29776

RESUMEN

En la actualidad se considera que se llega a un estado de malignidad a partir de una secuencia de pasos que se inician con la aparici[on de una c[elulas premaligna (portadora de una mutaci[on), la que desde ese momento se detiene en su estadio de diferenciaci[on, manteniendo su capacidad de divisi[on. La aparici[on posterior de cambios genéticos específicos sería responsable de la transición de células francamente malignas que poseen anomalías cromosómicas características. La obsrevación de anomalías cromosómicas específicas en células neoplásicas corrobora el papel fundamental que tienen estas anomalías en la transformación de una célula normal a otra maligna. Si bien no se conocen con exactitud los mecanismos por los cuales se originan las alteraciones cromosómicas, actualmente se acepta que pueden verse facilitadas por la presencia, entre otros, de los llamados sitios frágiles (SF), observables en los cromosomas (1). La primera mención de SF fue publicada en 1965 por Dekaban, describiendo un clon de células con un cromosoma del grupo C anorormal (2). Tres años más tarde, Lejeune, demostró la heredabilidad de estos sitios (3). En 1969, Lubs citó un SF en el cromosoma X (fra(X)) asociado al retardo mental (4). En los últimos tres años, se han llegado a identificar alrededor de 90 SF, de los cuales solamente el fra(X) está relacionado con una anormalidad fenotípica, mientras que se postula que los restnates pueden actuar como factores predisponentes a rupturas y a rearreglos cromosómicos, muchos de ellos, involucrados en diferentes neoplasias humanas (1). Esta idea se ve apoyada por el hecho de que los puntos de ruptura de ciertos reordenamientos estructurales coinciden con la ubicación cromosómica de determinados sitios frágiles (AU)


Asunto(s)
Humanos , Fragilidad Cromosómica , Neoplasias/genética , Fragilidad Cromosómica/efectos de los fármacos , Oncogenes , Aberraciones Cromosómicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...