Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Blood ; 114(14): 3001-7, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19666867

RESUMEN

Minimally differentiated acute myeloid leukemia (AML-M0) is defined by immature morphology and expression of early hematologic markers. By gene expression profiling (GEP) and subsequent unsupervised analysis of 35 AML-M0 samples and 253 previously reported AML cases, we demonstrate that AML-M0 cases express a unique signature that is largely separated from other molecular subtypes. Hematologic transcription regulators such as CEBPA, CEBPD, and ETV6, and the differentiation associated gene MPO appeared strongly down-regulated, in line with the primitive state of this leukemia. AML-M0 frequently carries loss-of-function RUNX1 mutation. Unsupervised analyses revealed a subdivision between AML-M0 cases with and without RUNX1 mutations. RUNX1 mutant AML-M0 samples showed a distinct up-regulation of B cell-related genes such as members of the B-cell receptor complex, transcription regulators RUNX3, ETS2, IRF8, or PRDM1, and major histocompatibility complex class II genes. Importantly, prediction with high accuracy of the AML-M0 subtype and prediction of patients carrying RUNX1 mutation within this subtype were possible based on the expression level of only a few transcripts. We propose that RUNX1 mutations in this AML subgroup cause lineage infidelity, leading to aberrant coexpression of myeloid and B-lymphoid genes. Furthermore, our results imply that AML-M0, although originally determined by morphology, constitutes a leukemia subgroup.


Asunto(s)
Biomarcadores de Tumor/genética , Diferenciación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/clasificación , Leucemia Mieloide Aguda/genética , Mutación/genética , Linfocitos B/patología , Redes Reguladoras de Genes , Humanos , Leucemia Mieloide Aguda/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Regulación hacia Arriba
2.
Haematologica ; 94(11): 1546-54, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19773259

RESUMEN

BACKGROUND: Minimally differentiated acute myeloid leukemia is heterogeneous in karyotype and is defined by immature morphological and molecular characteristics. This originally French-American-British classification is still used in the new World Health Organization classification when other criteria are not met. Apart from RUNX1 mutation, no characteristic molecular aberrations are recognized. DESIGN AND METHODS: We performed whole genome single nucleotide polymorphism analysis and extensive molecular analysis in a cohort of 52 patients with minimally differentiated acute myeloid leukemia. RESULTS: Many recurring and potentially relevant regions of loss of heterozygosity were revealed. These point towards a variety of candidate genes that could contribute to the pathogenesis of minimally differentiated acute myeloid leukemia, including the tumor suppressor genes TP53 and NF1, and reinforced the importance of RUNX1 in this leukemia. Furthermore, for the first time in this minimally differentiated form of leukemia we detected mutations in the transactivation domain of RUNX1. Mutations in other acute myeloid leukemia associated transcriptions factors were infrequent. In contrast, FLT3, RAS, PTPN11 and JAK2 were often mutated. Irrespective of the RUNX1 mutation status, our results show that RAS signaling is the most important pathway for proliferation in minimally differentiated acute myeloid leukemia. Importantly, we found that high terminal deoxynucleotidyl transferase expression is closely associated with RUNX1 mutation, which could allow an easier diagnosis of RUNX1 mutation in this hematologic malignancy. CONCLUSIONS: Our results suggest that in patients without RUNX1 mutation, several other molecular aberrations, separately or in combination, contribute to a common minimally differentiated phenotype.


Asunto(s)
Genoma Humano , Leucemia Mieloide Aguda/clasificación , Leucemia Mieloide Aguda/genética , Mutación , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Diferenciación Celular , Niño , Preescolar , Estudios de Cohortes , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Humanos , Cariotipificación , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Adulto Joven
3.
Mutat Res ; 663(1-2): 7-14, 2009 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-19150617

RESUMEN

Irradiation of cells with UVC light induces two types of mutagenic DNA photoproducts, i.e. cyclobutane pyrimidine dimers (CPD) and pyrimidine (6-4) pyrimidone photoproducts (6-4 PP). To investigate the relationship between the frequency of UV-induced photolesions at specific sites and their ability to induce mutations, we quantified CPD formation at the nucleotide level along exons 3 and 8 of the hprt gene using ligation-mediated PCR, and determined the mutational spectrum of 132 UV-induced hprt mutants in the AA8 hamster cell line and of 165 mutants in its nucleotide excision repair-defective derivative UV5. In AA8 cells, transversions predominated with a strong strand bias towards thymine-containing photolesions in the non-transcribed strand. As hamster AA8 cells are proficient in global genome repair of 6-4 PP but selectively repair CPD from the transcribed strand of active genes, most mutations probably resulted from erroneous bypass of CPD in the non-transcribed strand. However, the relative incidence of CPD and the positions where mutations most frequently arose do not correlate. In fact some major damage sites hardly gave rise to the formation of mutations. In the repair-defective UV5 cells, mutations were almost exclusively C>T transitions caused by photoproducts at PyC sites in the transcribed strand. Even though CPD were formed at high frequencies at some TT sites in UV5, these photoproducts did not contribute to mutation induction at all. We conclude that, even in the absence of repair, large variations in the level of induction of CPD at different sites throughout the two exons do not correspond to frequencies of mutation induction.


Asunto(s)
Reparación del ADN/efectos de la radiación , Mutación/genética , Dímeros de Pirimidina/metabolismo , Rayos Ultravioleta , Animales , Secuencia de Bases , Células CHO , Cricetinae , Cricetulus , Exones/genética , Hipoxantina Fosforribosiltransferasa/genética , Datos de Secuencia Molecular
4.
Haematologica ; 92(8): 1123-6, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17650443

RESUMEN

Of 52 AML-M0 patients studied, 16 presented a RUNX1 mutation (30.8 %) and 8 carried a trisomy 13 (15 %). We found a strong correlation between trisomy 13 and RUNX1 mutations, i.e, 7 out of 8 cases with trisomy 13 carried a mutation in RUNX1 (87.5 %, p<0.00056). Trisomy 13 patients with a RUNX1 mutation showed a 4-fold higher expression of FLT3 mRNA compared to controls, and in a selected number of cases, a higher cell fraction expressing FLT3 and an increase in the number of FLT3 receptors at the cell surface. In conclusion, our results show that trisomy 13 is correlated to RUNX1 mutation and increased FLT3 expression in AML-M0.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide/genética , Mutación , Proteínas de Neoplasias/genética , Trisomía , Tirosina Quinasa 3 Similar a fms/genética , Enfermedad Aguda , Estudios de Cohortes , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Exones/genética , Humanos , Cariotipificación , Leucemia Mieloide/clasificación , Leucemia Mieloide/metabolismo , Proteínas de Neoplasias/biosíntesis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Tirosina Quinasa 3 Similar a fms/biosíntesis
5.
Mutat Res ; 617(1-2): 58-70, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17327130

RESUMEN

Accumulation of damage in undifferentiated cells may threaten homeostasis and regenerative capacity. Remarkably, p53 has been suggested to be transcriptionally inactive in these cells. To gain insight in the kinetics and interplay of the predominant transcriptional responses of DNA damage signalling pathways in undifferentiated cells, mouse embryonic stem cells were exposed to cisplatin at four different time points (2, 4, 8 and 24h) and concentrations (1, 2, 5 and 10 microM). RNA was isolated and subjected to genome-wide expression profiling. Up to one fourth of the tested genes could be identified as being differentially expressed (false discovery rate=10%) after the cisplatin treatment. Clustering of the expression changes showed a strong time dependency. To investigate the relationship between affected genes, a gene set analysis method was used. Functionally related gene sets were defined using gene ontologies or transcription factor binding sites and were tested for overrepresentation within the differentially expressed genes. A variety of gene sets were clearly enriched among which 'apoptosis' and 'cell cycle' were the most pronounced. Furthermore, there was a strong enrichment of genes with a p53-binding motif. The involvement of the 'cell cycle' and 'apoptosis' gene sets in the cisplatin response was detected at concentrations and time points where the respective biological assays were still negative. The results reveal novel insights into the mechanisms which maintain the genomic integrity in undifferentiated cells. Additionally the results illustrate that gene set analysis of genome-wide expression changes provides a sensitive instrument to detect cellular stress responses to DNA damage.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Proteína p53 Supresora de Tumor/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
PLoS Med ; 3(10): e422, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17076557

RESUMEN

BACKGROUND: Radiation is an effective anti-cancer therapy but leads to severe late radiation toxicity in 5%-10% of patients. Assuming that genetic susceptibility impacts this risk, we hypothesized that the cellular response of normal tissue to X-rays could discriminate patients with and without late radiation toxicity. METHODS AND FINDINGS: Prostate carcinoma patients without evidence of cancer 2 y after curative radiotherapy were recruited in the study. Blood samples of 21 patients with severe late complications from radiation and 17 patients without symptoms were collected. Stimulated peripheral lymphocytes were mock-irradiated or irradiated with 2-Gy X-rays. The 24-h radiation response was analyzed by gene expression profiling and used for classification. Classification was performed either on the expression of separate genes or, to augment the classification power, on gene sets consisting of genes grouped together based on function or cellular colocalization.X-ray irradiation altered the expression of radio-responsive genes in both groups. This response was variable across individuals, and the expression of the most significant radio-responsive genes was unlinked to radiation toxicity. The classifier based on the radiation response of separate genes correctly classified 63% of the patients. The classifier based on affected gene sets improved correct classification to 86%, although on the individual level only 21/38 (55%) patients were classified with high certainty. The majority of the discriminative genes and gene sets belonged to the ubiquitin, apoptosis, and stress signaling networks. The apoptotic response appeared more pronounced in patients that did not develop toxicity. In an independent set of 12 patients, the toxicity status of eight was predicted correctly by the gene set classifier. CONCLUSIONS: Gene expression profiling succeeded to some extent in discriminating groups of patients with and without severe late radiotherapy toxicity. Moreover, the discriminative power was enhanced by assessment of functionally or structurally related gene sets. While prediction of individual response requires improvement, this study is a step forward in predicting susceptibility to late radiation toxicity.


Asunto(s)
Carcinoma/metabolismo , Carcinoma/radioterapia , Perfilación de la Expresión Génica/métodos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/radioterapia , Traumatismos por Radiación/diagnóstico , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Análisis por Conglomerados , Humanos , Linfocitos/metabolismo , Linfocitos/efectos de la radiación , Masculino , Análisis por Micromatrices , Persona de Mediana Edad , Modelos Biológicos , Dosis de Radiación , Traumatismos por Radiación/clasificación , Traumatismos por Radiación/metabolismo , Radiación Ionizante
7.
Oncogene ; 22(4): 538-47, 2003 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-12555067

RESUMEN

Based on our previous results indicating the presence of a tumor suppressor gene (TSG), chromosome 21 was analysed for loss of heterozygosity (LOH) in 18 patients with acute myeloid leukemia (17, AML-M0; one, AML-M1). Allelotyping at polymorphic loci was performed on purified material, allowing unequivocal detection of allelic loss and homozygous deletions. Six AML-M0 patients shared a common region of LOH harboring a single gene: RUNX1 (AML1), the most frequent site of translocations in acute leukemia and a well-known fusion oncogene. Fluorescence in situ hybridization allowed the identification of deletions with breakpoints within RUNX1 in two patients as the cause of LOH. In the four others the LOH pattern and the presence of two karyotypically normal chromosomes 21 were in line with mitotic recombination. Further molecular and cytogenetic analyses showed that this caused homozygosity of primary RUNX1 mutations: two point mutations, a partial deletion and, most significantly, a complete deletion of RUNX1. These findings identify RUNX1 as a classical TSG: both alleles are mutated or absent in cancer cells from four of the 17 AML-M0 patients examined. In contrast to AML-M0, the AML-M1 patient was trisomic for chromosome 21 and has two mutated and one normal RUNX1 allele, suggesting that the order of mutagenic events leading to leukemia may influence the predominant tumor type.


Asunto(s)
Proteínas de Unión al ADN/genética , Genes Supresores de Tumor , Proteínas Proto-Oncogénicas , Factores de Transcripción/genética , Enfermedad Aguda , Cromosomas Humanos Par 21 , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Citometría de Flujo , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Leucemia Mieloide/genética , Pérdida de Heterocigocidad , Mutación Puntual
8.
BMC Bioinformatics ; 6: 177, 2005 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-16018805

RESUMEN

BACKGROUND: Functional analysis of data from genome-scale experiments, such as microarrays, requires an extensive selection of differentially expressed genes. Under many conditions, the proportion of differentially expressed genes is considerable, making the selection criteria a balance between the inclusion of false positives and the exclusion of false negatives. RESULTS: We developed an analytical method to determine a p-value threshold from a microarray experiment that is dependent on the quality and design of the data set. To this aim, populations of p-values are modeled as mathematical functions in which the parameters to describe these functions are estimated in an unsupervised manner. The strength of the method is exemplified by its application to a published gene expression data set of sporadic and familial breast tumors with BRCA1 or BRCA2 mutations. CONCLUSION: We present an objective and unsupervised way to set thresholds adapted to the quality and design of the experiment. The resulting mathematical description of the data sets of genome-scale experiments enables a probabilistic approach in systems biology.


Asunto(s)
Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Modelos Genéticos , Neoplasias de la Mama/genética , Ciclo Celular/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Pruebas Genéticas/métodos , Humanos , Fosforilación , Valor Predictivo de las Pruebas , Análisis por Matrices de Proteínas/métodos
9.
Radiat Res ; 177(5): 602-13, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22468706

RESUMEN

The recent steep increase in population dose from radiation-based medical diagnostics, such as computed tomography (CT) scans, requires insight into human health risks, especially in terms of cancer development. Since the induction of genetic damage is considered a prominent cause underlying the carcinogenic potential of ionizing radiation, we quantified the induction of micronuclei and loss of heterozygosity events in human cells after exposure to clinically relevant low doses of X rays. A linear dose-response relationship for induction of micronuclei was observed in human fibroblasts with significantly increased frequencies at doses as low as 20 mGy. Strikingly, cells exposed during S-phase displayed the highest induction, whereas non S-phase cells showed no significant induction below 100 mGy. Similarly, the induction of loss of heterozygosity in human lymphoblastoid cells quantified at HLA loci, was linear with dose and reached significance at 50 mGy. Together the findings favor a linear-no-threshold model for genetic damage induced by acute exposure to ionizing radiation. We speculate that the higher radiosensitivity of S-phase cells might relate to the excessive cancer risk observed in highly proliferative tissues in radiation exposed organisms.


Asunto(s)
Aberraciones Cromosómicas , Cromosomas Humanos/efectos de la radiación , Linfocitos/efectos de la radiación , Rayos X/efectos adversos , División Celular/efectos de la radiación , Células Cultivadas/efectos de la radiación , Células Cultivadas/ultraestructura , Relación Dosis-Respuesta en la Radiación , Fibroblastos/efectos de la radiación , Fibroblastos/ultraestructura , Genes MHC Clase I/efectos de la radiación , Humanos , Pérdida de Heterocigocidad , Linfocitos/ultraestructura , Pruebas de Micronúcleos , Tolerancia a Radiación , Radiografía , Reproducibilidad de los Resultados , Fase S/efectos de la radiación
10.
J Pathol ; 208(4): 564-73, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16353169

RESUMEN

Approximately 60% of sporadic meningiomas are caused by inactivation of the NF2 tumour suppressor gene. The causative gene for the remaining meningiomas is unknown. Previous studies have shown that these tumours have no recurrent karyotypic abnormalities. They differ from their NF2-related counterparts in that they are more often of the meningothelial subtype and are located preferentially in the anterior skull base. To gain more insight into the aetiology of these tumours, we studied genetic and epigenetic alterations in 25 meningiomas without NF2 involvement. We first established a genome-wide allelotype using 3 microsatellite markers per chromosome arm. Loss of heterozygosity (LOH) was detected at a low frequency and no indication for the location of putative tumour suppressor genes could be established. We next screened the subtelomeric regions by using 2-3 polymorphic markers close to each telomere. Again no evidence for LOH of a particular chromosome arm was obtained, and no LOH was found in the genomic regions containing the NF2-related ERM family members ezrin and radixin, DAL-1, protein 4.1R, and TSLC1. Mutations in the X-chromosome based family member, moesin, were analysed by SSCP and were not detected. Microsatellite instability was studied using 6 commonly used markers but none of these was altered in any meningioma. Methylation was detected in 5 of 16 genes (NF2, p14(ARF), CDH1, BRCA1, RB1) previously shown to be silenced in a variety of tumour types. However, methylation percentages for these genes were generally higher in a group of NF2-related meningiomas, with the exception of the BRCA1 gene. The NF2 gene was methylated in only 1 of 21 tumours. In conclusion, meningiomas with an intact NF2 gene have a normal karyotype and no obvious genetic or epigenetic aberrations, suggesting that the gene(s) involved in the pathogenesis of these tumours are altered by smaller events than can be detected with the techniques used in our study.


Asunto(s)
Meningioma/genética , Proteínas de Neoplasias/genética , Neoplasias de la Base del Cráneo/genética , Adulto , Anciano , Alelos , Metilación de ADN , Análisis Mutacional de ADN , Femenino , Genes BRCA1 , Marcadores Genéticos , Humanos , Pérdida de Heterocigocidad , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Neurofibromina 2/genética , Polimorfismo Conformacional Retorcido-Simple
11.
Genes Chromosomes Cancer ; 35(1): 38-48, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12203788

RESUMEN

Loss of heterozygosity (LOH) is a major mechanism for inactivation of tumor-suppressor genes and has been observed in various solid tumors and lymphomas. The human leukocyte antigen (HLA) region is located at chromosome band 6p21.3, and loss or alteration of this region may provide tumor cells with a mechanism to escape from the immune system. We previously identified small homozygous deletions within the HLA class II region in many of the diffuse large B-cell lymphomas (DLCLs) of the central nervous system (CNS) and the testis. In the present study, we focused on the mechanism leading to LOH in the HLA region. Twenty microsatellite markers, of which 12 were specific for HLA, were applied on 11 extranodal DLCLs of the CNS and 28 of the testis. Additionally, fluorescence in situ hybridization with seven HLA-specific probes and a centromere 6-specific probe was performed on 20 cases to study the mechanism of LOH. In contrast to previously published data on spontaneously mutated lymphoblastoid cell lines, intrachromosomal hemizygous deletion, not mitotic recombination, was the major cause of LOH of the HLA region in these lymphomas. However, opposed to data in colorectal cancer, these deletions were rarely (one of nine cases) associated with an interchromosomal rearrangement such as a translocation.


Asunto(s)
Deleción Cromosómica , Antígenos HLA/genética , Pérdida de Heterocigocidad/genética , Linfoma de Células B/genética , Linfoma de Células B Grandes Difuso/genética , Neoplasias Testiculares/genética , Alelos , Inversión Cromosómica , Segregación Cromosómica/genética , Cromosomas Humanos Par 6/genética , Sondas de ADN/genética , Humanos , Hibridación Fluorescente in Situ/métodos , Interfase/genética , Masculino , Mitosis/genética , Monosomía/genética , Recombinación Genética/genética , Translocación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA