Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Haematologica ; 99(5): 848-57, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24441149

RESUMO

Acute myeloid leukemia is a neoplasm characterized by recurrent molecular aberrations traditionally demonstrated by cytogenetic analyses. We used high density genome-wide genotyping and gene expression profiling to reveal acquired cryptic abnormalities in acute myeloid leukemia. By genome-wide genotyping of 137 cases of primary acute myeloid leukemia, we disclosed a recurrent focal amplification on chromosome 14q32, which included the genes BCL11B, CCNK, C14orf177 and SETD3, in two cases. In the affected cases, the BCL11B gene showed consistently high mRNA expression, whereas the expression of the other genes was unperturbed. Fluorescence in situ hybridization on 40 cases of acute myeloid leukemia with high BCL11B mRNA expression [2.5-fold above median; 40 out of 530 cases (7.5%)] revealed 14q32 abnormalities in two additional cases. In the four BCL11B-rearranged cases the 14q32 locus was fused to different partner chromosomes. In fact, in two cases, we demonstrated that the focal 14q32 amplifications were integrated into transcriptionally active loci. The translocations involving BCL11B result in increased expression of full-length BCL11B protein. The BCL11B-rearranged acute myeloid leukemias expressed both myeloid and T-cell markers. These biphenotypic acute leukemias all carried FLT3 internal tandem duplications, a characteristic marker of acute myeloid leukemia. BCL11B mRNA expression in acute myeloid leukemia appeared to be strongly associated with expression of other T-cell-specific genes. Myeloid 32D(GCSF-R) cells ectopically expressing Bcl11b showed decreased proliferation rate and less maturation. In conclusion, by an integrated approach involving high-throughput genome-wide genotyping and gene expression profiling we identified BCL11B as a candidate oncogene in acute myeloid leukemia.


Assuntos
Aberrações Cromossômicas , Cromossomos Humanos Par 14 , Leucemia Mieloide Aguda/genética , Oncogenes , Proteínas Repressoras/genética , Proteínas Supressoras de Tumor/genética , Antígenos de Superfície/metabolismo , Estudos de Casos e Controles , Proliferação de Células , Dosagem de Genes , Expressão Gênica , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Imunofenotipagem , Hibridização in Situ Fluorescente , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/metabolismo , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA , Linfócitos T/metabolismo , Translocação Genética
2.
Genesis ; 47(1): 7-13, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18942097

RESUMO

To develop a sensitive and inducible system to study intestinal biology, we generated a transgenic mouse model expressing the reverse tetracycline transactivator rtTA2-M2 under control of the 12.4 kb murine Villin promoter. The newly generated Villin-rtTA2-M2 mice were then bred with the previously developed tetO-HIST1H2BJ/GFP model to assess inducibility and tissue-specificity. Expression of the histone H2B-GFP fusion protein was observed exclusively upon doxycycline induction and was uniformly distributed throughout the intestinal epithelium. The Villin-rtTA2-M2 was also found to drive transgene expression in the developing mouse intestine. Furthermore, we could detect transgene expression in the proximal tubules of the kidney and in a population of alleged gastric progenitor cells. By administering different concentrations of doxycycline, we show that the Villin-rtTA2-M2 system drives transgene expression in a dosage-dependent fashion. Thus, we have generated a novel doxycycline-inducible mouse model, providing a valuable tool to study the effect of different gene dosages on intestinal physiology and pathology.


Assuntos
Doxiciclina/farmacologia , Expressão Gênica/efeitos dos fármacos , Engenharia Genética/métodos , Intestinos/fisiologia , Animais , Genes Reporter/genética , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos , Pesquisa , Titulometria , Transgenes/genética
3.
Clin Cancer Res ; 14(12): 3916-25, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18559613

RESUMO

PURPOSE: To assess efficacy of the novel, selective poly(ADP-ribose) polymerase-1 (PARP-1) inhibitor AZD2281 against newly established BRCA2-deficient mouse mammary tumor cell lines and to determine potential synergy between AZD2281 and cisplatin. EXPERIMENTAL DESIGN: We established and thoroughly characterized a panel of clonal cell lines from independent BRCA2-deficient mouse mammary tumors and BRCA2-proficient control tumors. Subsequently, we assessed sensitivity of these lines to conventional cytotoxic drugs and the novel PARP inhibitor AZD2281. Finally, in vitro combination studies were done to investigate interaction between AZD2281 and cisplatin. RESULTS: Genetic, transcriptional, and functional analyses confirmed the successful isolation of BRCA2-deficient and BRCA2-proficient mouse mammary tumor cell lines. Treatment of these cell lines with 11 different anticancer drugs or with gamma-irradiation showed that AZD2281, a novel and specific PARP inhibitor, caused the strongest differential growth inhibition of BRCA2-deficient versus BRCA2-proficient mammary tumor cells. Finally, drug combination studies showed synergistic cytotoxicity of AZD2281 and cisplatin against BRCA2-deficient cells but not against BRCA2-proficient control cells. CONCLUSION: We have successfully established the first set of BRCA2-deficient mammary tumor cell lines, which form an important addition to the existing preclinical models for BRCA-mutated breast cancer. The exquisite sensitivity of these cells to the PARP inhibitor AZD2281, alone or in combination with cisplatin, provides strong support for AZD2281 as a novel targeted therapeutic against BRCA-deficient cancers.


Assuntos
Proteína BRCA2/genética , Proliferação de Células/efeitos dos fármacos , Cisplatino/administração & dosagem , Neoplasias Mamárias Animais/tratamento farmacológico , Ftalazinas/administração & dosagem , Piperazinas/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Proteína BRCA2/metabolismo , Linhagem Celular Tumoral , Dano ao DNA , Avaliação Pré-Clínica de Medicamentos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores Enzimáticos/farmacologia , Feminino , Neoplasias Mamárias Animais/genética , Camundongos , Camundongos Transgênicos , Células-Tronco Neoplásicas/efeitos da radiação , Inibidores de Poli(ADP-Ribose) Polimerases , Rad51 Recombinase/genética
4.
Genes Chromosomes Cancer ; 47(3): 185-96, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18050305

RESUMO

Testicular germ cell tumors of adolescents and adults (TGCTs) can be classified into seminomatous and nonseminomatous tumors. Various nonseminomatous cell lines, predominantly embryonal carcinoma, have been established and proven to be valuable for pathobiological and clinical studies. So far, no cell lines have been derived from seminoma which constitutes more than 50% of invasive TGCTs. Such a cell line is essential for experimental investigation of biological characteristics of the cell of origin of TGCTs, i.e., carcinoma in situ of the testis, which shows characteristics of a seminoma cell. Before a cell line can be used as model, it must be verified regarding its origin and characteristics. Therefore, a multidisciplinary approach was undertaken on TCam-2 cells, supposedly the first seminoma cell line. Fluorescence in situ hybridization, array comparative genomic hybridization, and spectral karyotyping demonstrated an aneuploid DNA content, with gain of 12p, characteristic for TGCTs. Genome wide mRNA and microRNA expression profiling supported the seminoma origin, in line with the biallelic expression of imprinted genes IGF2/H19 and associated demethylation of the imprinting control region. Moreover, the presence of specific markers, demonstrated by immunohistochemistry, including (wild type) KIT, stem cell factor, placental alkaline phosphatase, OCT3/4 (also demonstrated by a specific Q-PCR) and NANOG, and the absence of CD30, SSX2-4, and SOX2, confirms that TCam-2 is a seminoma cell line. Although mutations in oncogenes and tumor suppressor genes are rather rare in TGCTs, TCam-2 had a mutated BRAF gene (V600E), which likely explains the fact that these cells could be propagated in vitro. In conclusion, TCam-2 is the first well-characterized seminoma-derived cell line, with an exceptional mutation, rarely found in TGCTs.


Assuntos
Linhagem Celular Tumoral , Seminoma/patologia , Perfilação da Expressão Gênica , Impressão Genômica , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Masculino , Proteínas de Neoplasias/análise , Hibridização de Ácido Nucleico , Cariotipagem Espectral
5.
Mol Biol Cell ; 16(2): 769-75, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15574874

RESUMO

The position of chromosomal neighborhoods in living cells was followed using three different methods for marking chromosomal domains occupying arbitrary locations in the nucleus; photobleaching of GFP-labeled histone H2B, local UV-marked DNA, and photobleaching of fluorescently labeled DNA. All methods revealed that global chromosomal organization can be reestablished through one cell division from mother to daughters. By simultaneously monitoring cell cycle stage in the cells in which relative chromosomal domain positions were tracked, we observed that chromosomal neighborhood organization is apparently lost in the early G1 phase of the cell cycle. However, the daughter cells eventually regain the general chromosomal organization pattern of their mothers, suggesting an active mechanism could be at play to reestablish chromosomal neighborhoods.


Assuntos
Ciclo Celular/genética , Cromossomos Humanos/metabolismo , Cromossomos de Mamíferos/metabolismo , Animais , Biomarcadores/metabolismo , Células CHO , Núcleo Celular/genética , Cromossomos Humanos/genética , Cromossomos de Mamíferos/genética , Células Clonais , Cricetinae , Cricetulus , DNA/metabolismo , DNA/efeitos da radiação , Dano ao DNA , DNA de Neoplasias/metabolismo , DNA de Neoplasias/efeitos da radiação , Técnica Indireta de Fluorescência para Anticorpo , Corantes Fluorescentes , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Hidrazinas , Cinética , Microscopia Confocal , Microscopia de Vídeo , Fotodegradação , Antígeno Nuclear de Célula em Proliferação/metabolismo , Transfecção , Raios Ultravioleta
6.
Cancer Res ; 66(1): 290-302, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16397242

RESUMO

Spermatocytic seminomas are solid tumors found solely in the testis of predominantly elderly individuals. We investigated these tumors using a genome-wide analysis for structural and numerical chromosomal changes through conventional karyotyping, spectral karyotyping, and array comparative genomic hybridization using a 32 K genomic tiling-path resolution BAC platform (confirmed by in situ hybridization). Our panel of five spermatocytic seminomas showed a specific pattern of chromosomal imbalances, mainly numerical in nature (range, 3-24 per tumor). Gain of chromosome 9 was the only consistent anomaly, which in one case also involved amplification of the 9p21.3-pter region. Parallel chromosome level expression profiling as well as microarray expression analyses (Affymetrix U133 plus 2.0) was also done. Unsupervised cluster analysis showed that a profile containing transcriptional data on 373 genes (difference of > or = 3.0-fold) is suitable for distinguishing these tumors from seminomas/dysgerminomas. The diagnostic markers SSX2-4 and POU5F1 (OCT3/OCT4), previously identified by us, were among the top discriminatory genes, thereby validating the experimental set-up. In addition, novel discriminatory markers suitable for diagnostic purposes were identified, including Deleted in Azospermia (DAZ). Although the seminomas/dysgerminomas were characterized by expression of stem cell-specific genes (e.g., POU5F1, PROM1/CD133, and ZFP42), spermatocytic seminomas expressed multiple cancer testis antigens, including TSP50 and CTCFL (BORIS), as well as genes known to be expressed specifically during prophase meiosis I (TCFL5, CLGN, and LDHc). This is consistent with different cells of origin, the primordial germ cell and primary spermatocyte, respectively. Based on the region of amplification defined on 9p and the associated expression plus confirmatory immunohistochemistry, DMRT1 (a male-specific transcriptional regulator) was identified as a likely candidate gene for involvement in the development of spermatocytic seminomas.


Assuntos
Cromossomos Humanos Par 9/genética , Seminoma/genética , Neoplasias Testiculares/genética , Fatores de Transcrição/genética , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Feminino , Perfilação da Expressão Gênica , Instabilidade Genômica , Humanos , Imuno-Histoquímica , Masculino , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Seminoma/metabolismo , Espermatócitos/patologia , Neoplasias Testiculares/metabolismo
7.
Mol Cell Biol ; 24(13): 5776-87, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15199134

RESUMO

Interstrand cross-links (ICLs) are an extremely toxic class of DNA damage incurred during normal metabolism or cancer chemotherapy. ICLs covalently tether both strands of duplex DNA, preventing the strand unwinding that is essential for polymerase access. The mechanism of ICL repair in mammalian cells is poorly understood. However, genetic data implicate the Ercc1-Xpf endonuclease and proteins required for homologous recombination-mediated double-strand break (DSB) repair. To examine the role of Ercc1-Xpf in ICL repair, we monitored the phosphorylation of histone variant H2AX (gamma-H2AX). The phosphoprotein accumulates at DSBs, forming foci that can be detected by immunostaining. Treatment of wild-type cells with mitomycin C (MMC) induced gamma-H2AX foci and increased the amount of DSBs detected by pulsed-field gel electrophoresis. Surprisingly, gamma-H2AX foci were also induced in Ercc1(-/-) cells by MMC treatment. Thus, DSBs occur after cross-link damage via an Ercc1-independent mechanism. Instead, ICL-induced DSB formation required cell cycle progression into S phase, suggesting that DSBs are an intermediate of ICL repair that form during DNA replication. In Ercc1(-/-) cells, MMC-induced gamma-H2AX foci persisted at least 48 h longer than in wild-type cells, demonstrating that Ercc1 is required for the resolution of cross-link-induced DSBs. MMC triggered sister chromatid exchanges in wild-type cells but chromatid fusions in Ercc1(-/-) and Xpf mutant cells, indicating that in their absence, repair of DSBs is prevented. Collectively, these data support a role for Ercc1-Xpf in processing ICL-induced DSBs so that these cytotoxic intermediates can be repaired by homologous recombination.


Assuntos
Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/genética , Endonucleases/genética , Animais , Ciclo Celular , Linhagem Celular , Aberrações Cromossômicas , DNA/química , DNA/metabolismo , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/fisiologia , Endonucleases/deficiência , Endonucleases/fisiologia , Raios gama , Histonas/análise , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Mitomicina/farmacologia , Conformação de Ácido Nucleico , Raios Ultravioleta
8.
Mutat Res ; 596(1-2): 22-35, 2006 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-16472827

RESUMO

Genetic defects in nucleotide excision repair (NER) are associated with premature aging, including cancer, in both humans and mice. To investigate the possible role of increased somatic mutation accumulation in the accelerated appearance of symptoms of aging as a consequence of NER deficiency, we crossed four different mouse mutants, Xpa-/-, Ercc6(Csb)-/-, Ercc2(Xpd)m/m and Ercc1-/m, with mice harboring lacZ-reporter genes to assess mutant frequencies and spectra in different organs during aging. The results indicate an accelerated accumulation of mutations in both liver and kidney of Xpa defective mice, which correlated with a trend towards a decreased lifespan. Until 52 weeks, Xpa deficiency resulted mainly in 1-bp deletions. At old age (104 weeks), the spectrum had undergone a shift, in both organs, to G:C-->T:A transversions, a signature mutation of oxidative DNA damage. Ercc1-/m mice, with their short lifespan of 6 months and severe symptoms of premature aging, especially in liver and kidney, displayed an even faster lacZ-mutant accumulation in liver. In this case, the excess mutations were mostly genome rearrangements. Csb-/- mice, with mild premature aging features and no reduction in lifespan, and Xpdm/m mice, exhibiting prominent premature aging features and about 20% reduction in lifespan, did not have elevated lacZ-mutant frequencies. It is concluded that while increased genomic instability could play a causal role in the mildly accelerated aging phenotype in the Xpa-null mice or in the severe progeroid symptoms of the Ercc1-mutant mice, shortened lifespan in mice with defects in transcription-related repair do not depend upon increased mutation accumulation.


Assuntos
Reparo do DNA/genética , Genoma , Mutação , Senilidade Prematura/genética , Animais , Sequência de Bases , Mapeamento Cromossômico , Cruzamentos Genéticos , Primers do DNA , Rearranjo Gênico , Genes Reporter , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , beta-Galactosidase/genética
9.
Clin Cancer Res ; 8(2): 534-9, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11839674

RESUMO

PURPOSE: In uveal melanoma, specific chromosomal abnormalities are known to correlate with the risk of metastases; changes in chromosomes 3 and 8q correlate strongly with a decreased survival of the patient, whereas chromosome 6 abnormalities are associated with a better prognosis. Usually, karyotyping and fluorescence in situ hybridization (FISH) analysis are used to detect these abnormalities in resected tumor tissues. However, the evaluation of these chromosomal changes is compromised in patients treated with eye-retaining treatment protocols because of the lack of tumor material. The purpose of this study was to validate the use of FISH for the analysis of genetic prognostic markers. EXPERIMENTAL DESIGN: We analyzed 40 uveal melanoma fine needle aspiration biopsies (FNABs) and the corresponding main tumor with FISH. RESULTS: All biopsies were found to contain tumor cells, and FISH analyses of the samples were successful in all cases. Statistical analysis showed very good agreement between the FISH results from the biopsies and those from the main tumor. In only 2 of 249 hybridizations did we find a small variation that could have led to a misclassification. CONCLUSIONS: Our results indicate that the application of FISH to FNABs is a reliable method for assaying genetic prognostic parameters such as chromosome 3 loss and/or chromosome 8q gain. Implementation of this method in a diagnostic setting means that we are able to identify patients at risk of developing metastatic disease, not only in enucleated patients but also in cases treated with conservative treatment modalities such as radiotherapy.


Assuntos
Hibridização in Situ Fluorescente/métodos , Melanoma/genética , Melanoma/patologia , Prognóstico , Neoplasias Uveais/genética , Neoplasias Uveais/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biópsia , Aberrações Cromossômicas , Cromossomos Humanos Par 3 , Cromossomos Humanos Par 6 , Cromossomos Humanos Par 8 , Feminino , Humanos , Cariotipagem , Masculino , Pessoa de Meia-Idade
10.
Cancer Cell ; 19(2): 244-56, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21316603

RESUMO

Small cell lung cancer (SCLC) is the lung neoplasia with the poorest prognosis, due to its high metastatic potential and chemoresistance upon relapse. Using the previously described mouse model for SCLC, we found that the tumors are often composed of phenotypically different cells with either a neuroendocrine or a mesenchymal marker profile. These cells had a common origin because they shared specific genomic aberrations. The transition from neuroendocrine to mesenchymal phenotype could be achieved by the ectopic expression of oncogenic Ras(V12). Crosstalk between mesenchymal and neuroendocrine cells strongly influenced their behavior. When engrafted as a mixed population, the mesenchymal cells endowed the neuroendocrine cells with metastatic capacity, illustrating the potential relevance of tumor cell heterogeneity in dictating tumor properties.


Assuntos
Carcinoma de Células Pequenas/patologia , Modelos Animais de Doenças , Neoplasias Pulmonares/patologia , Animais , Carcinoma de Células Pequenas/genética , Linhagem Celular Tumoral , Técnicas de Cocultura , Genes ras , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/genética , Camundongos , Metástase Neoplásica
11.
DNA Repair (Amst) ; 10(11): 1095-105, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21885354

RESUMO

Ultraviolet (UV) radiation-induced DNA lesions can be efficiently repaired by nucleotide excision repair (NER). However, NER is less effective during replication of UV-damaged chromosomes. In contrast, translesion DNA synthesis (TLS) and homologous recombination (HR) are capable of dealing with lesions in replicating DNA. The core HR protein in mammalian cells is the strand exchange protein RAD51, which is aided by numerous proteins, including RAD54. We used RAD54 as a cellular marker for HR to study the response of mammalian embryonic stem (ES) cells to UV irradiation. In contrast to yeast, ES cells lacking RAD54 are not UV sensitive. Here we show that the requirement for mammalian RAD54 is masked by active NER. By genetically inactivating NER and HR through disruption of the Xpa and Rad54 genes, respectively, we demonstrate the contribution of HR to chromosomal integrity upon UV irradiation. We demonstrate using chromosome fiber analysis at the individual replication fork level, that HR activity is important for the restart of DNA replication after induction of DNA damage by UV-light in NER-deficient cells. Furthermore, our data reveal RAD54-dependent and -independent contributions of HR to the cellular sensitivity to UV-light, and they uncover that RAD54 can compensate for the loss of TLS polymerase η with regard to UV-light sensitivity. In conclusion, we show that HR is important for the progression of UV-stalled replication forks in ES cells, and that protection of the fork is an interplay between HR and TLS.


Assuntos
Recombinação Homóloga/efeitos da radiação , Proteínas Nucleares/metabolismo , Raios Ultravioleta , Animais , Sobrevivência Celular/efeitos da radiação , Aberrações Cromossômicas/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos da radiação , DNA Helicases , Replicação do DNA , Proteínas de Ligação a DNA , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/efeitos da radiação , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Transporte Proteico/efeitos da radiação , Transdução de Sinais/efeitos da radiação
12.
DNA Repair (Amst) ; 8(12): 1363-70, 2009 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-19782649

RESUMO

Nijmegen breakage syndrome (NBS) is characterized by genome instability and cancer predisposition. NBS patients contain a mutation in the NBS1 gene, which encodes the NBS1 component of the DNA double-strand break (DSB) response complex MRE11/RAD50/NBS1. To investigate the NBS phenotype in more detail, we combined the mouse mimic of the most common patient mutation (Nbs1(Delta B/DeltaB)) with a Rad54 null mutation, which diminishes homologous recombination. Double mutant cells were particularly sensitive to treatments that cause single strand breaks (SSBs), presumably because these SSBs can be converted into detrimental DSBs upon passage of a replication fork. The persistent presence of nuclear RAD51 foci and increased levels of chromatid type breaks in metaphase spreads indicated that replication-associated DSBs are repaired inefficiently in the double mutant cells. We conclude that Nbs1 and Rad54 function cooperatively, but in separate pathways to counteract this type of DNA damage and discuss mechanistic implications of these findings.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Quebra Cromossômica , Dano ao DNA , Replicação do DNA , DNA/genética , Proteínas Nucleares/metabolismo , Recombinação Genética , Animais , Ciclo Celular , Proteínas de Ciclo Celular/genética , Células Cultivadas , DNA Helicases , Proteínas de Ligação a DNA , Feminino , Camundongos , Síndrome de Quebra de Nijmegen/genética , Síndrome de Quebra de Nijmegen/metabolismo , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética
13.
Blood ; 111(8): 4329-37, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18272813

RESUMO

Inappropriate expression of EVI1 (ecotropic virus integration-1), in particular splice form EVI1-1D, through chromosome 3q26 lesions or other mechanisms has been implicated in the development of high-risk acute myeloid leukemia (AML). To validate the clinical relevance of EVI1-1D, as well as of the other EVI1 splice forms and the related MDS1/EVI1 (ME) gene, real-time quantitative polymerase chain reaction was performed in 534 untreated adults with de novo AML. EVI1-1D was highly expressed in 6% of cases (n = 32), whereas 7.8% were EVI1(+) (n = 41) when all splice variants were taken into account. High EVI1 predicted a distinctly worse event-free survival (HR = 1.9; P = .002) and disease-free survival (HR = 2.1, P = .006) following multivariate analysis. Importantly, we distinguished a subset of EVI1(+) cases that lacked expression of ME (EVI1(+)ME(-); n = 17) from cases that were ME(+) (EVI1(+)ME(+); n = 24). The atypical EVI1(+)ME(-) expression pattern exhibited cytogenetically detectable chromosomal 3q26 breakpoints in 8 cases. Fluorescence in situ hybridization revealed 7 more EVI1(+)ME(-) cases that carried cryptic 3q26 breakpoints, which were not found in the EVI1(+)ME(+) group. EVI1(+)ME(-) expression predicts an extremely poor prognosis distinguishable from the general EVI1(+) AML patients (overall survival [OS]: P < .001 and event-free survival [EFS]: P = .002). We argue that EVI1/ME quantitative expression analysis should be implemented in the molecular diagnostic procedures of AML.


Assuntos
Aberrações Cromossômicas , Cromossomos Humanos Par 3/genética , Proteínas de Ligação a DNA/genética , Leucemia Mieloide Aguda/patologia , Proto-Oncogenes/genética , Fatores de Transcrição/genética , Adulto , Idoso , Processamento Alternativo/genética , Estudos de Coortes , Análise Citogenética , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação Leucêmica da Expressão Gênica , Humanos , Hibridização in Situ Fluorescente , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Proteína do Locus do Complexo MDS1 e EVI1 , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Prognóstico , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reprodutibilidade dos Testes , Análise de Sobrevida , Fatores de Transcrição/metabolismo , Resultado do Tratamento
14.
Mol Cell Biol ; 28(16): 5082-92, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18541667

RESUMO

ERCC1-XPF endonuclease is required for nucleotide excision repair (NER) of helix-distorting DNA lesions. However, mutations in ERCC1 or XPF in humans or mice cause a more severe phenotype than absence of NER, prompting a search for novel repair activities of the nuclease. In Saccharomyces cerevisiae, orthologs of ERCC1-XPF (Rad10-Rad1) participate in the repair of double-strand breaks (DSBs). Rad10-Rad1 contributes to two error-prone DSB repair pathways: microhomology-mediated end joining (a Ku86-independent mechanism) and single-strand annealing. To determine if ERCC1-XPF participates in DSB repair in mammals, mutant cells and mice were screened for sensitivity to gamma irradiation. ERCC1-XPF-deficient fibroblasts were hypersensitive to gamma irradiation, and gammaH2AX foci, a marker of DSBs, persisted in irradiated mutant cells, consistent with a defect in DSB repair. Mutant mice were also hypersensitive to irradiation, establishing an essential role for ERCC1-XPF in protecting against DSBs in vivo. Mice defective in both ERCC1-XPF and Ku86 were not viable. However, Ercc1(-/-) Ku86(-/-) fibroblasts were hypersensitive to gamma irradiation compared to single mutants and accumulated significantly greater chromosomal aberrations. Finally, in vitro repair of DSBs with 3' overhangs led to large deletions in the absence of ERCC1-XPF. These data support the conclusion that, as in yeast, ERCC1-XPF facilitates DSB repair via an end-joining mechanism that is Ku86 independent.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Endonucleases/metabolismo , Animais , Antígenos Nucleares/metabolismo , Linhagem Celular Transformada , Sobrevivência Celular/efeitos da radiação , Senescência Celular/efeitos da radiação , Aberrações Cromossômicas/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos da radiação , Proteínas de Ligação a DNA/deficiência , Perda do Embrião/metabolismo , Embrião de Mamíferos/citologia , Endonucleases/deficiência , Fibroblastos/enzimologia , Fibroblastos/efeitos da radiação , Instabilidade Genômica/efeitos da radiação , Células HeLa , Histonas/metabolismo , Humanos , Autoantígeno Ku , Camundongos , Plasmídeos/genética , Radiação Ionizante , Análise de Sequência de DNA
15.
Nat Struct Mol Biol ; 14(11): 1096-104, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17934473

RESUMO

Faithful duplication of the genome requires structure-specific endonucleases such as the RuvABC complex in Escherichia coli. These enzymes help to resolve problems at replication forks that have been disrupted by DNA damage in the template. Much less is known about the identities of these enzymes in mammalian cells. Mus81 is the catalytic component of a eukaryotic structure-specific endonuclease that preferentially cleaves branched DNA substrates reminiscent of replication and recombination intermediates. Here we explore the mechanisms by which Mus81 maintains chromosomal stability. We found that Mus81 is involved in the formation of double-strand DNA breaks in response to the inhibition of replication. Moreover, in the absence of chromosome processing by Mus81, recovery of stalled DNA replication forks is attenuated and chromosomal aberrations arise. We suggest that Mus81 suppresses chromosomal instability by converting potentially detrimental replication-associated DNA structures into intermediates that are more amenable to DNA repair.


Assuntos
Cromossomos de Mamíferos/química , Cromossomos de Mamíferos/metabolismo , Quebras de DNA de Cadeia Dupla , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Endonucleases/metabolismo , Conformação de Ácido Nucleico , Animais , Afidicolina/farmacologia , Ciclo Celular/fisiologia , Células Cultivadas , Instabilidade Cromossômica , Aberrações Cromossômicas/induzido quimicamente , Cromossomos de Mamíferos/genética , DNA Helicases , Proteínas de Ligação a DNA/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/efeitos da radiação , Endonucleases/genética , Inibidores Enzimáticos/farmacologia , Humanos , Hidroxiureia/farmacologia , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Inibidores da Síntese de Ácido Nucleico/farmacologia , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Radiação Ionizante
16.
Genes Chromosomes Cancer ; 46(10): 895-908, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17620293

RESUMO

Malignant mesothelioma (MM) is an asbestos-induced tumor that acquires aneuploid DNA content during the tumorigenic process. We used instable MM cell lines as an in vitro model to study the impact of DNA copy-number changes on gene expression profiling, in the course of their chromosomal redistribution process. Two MM cell lines, PMR-MM2 (early passages of in vitro culture) and PMR-MM7 (both early and late passages of in vitro culture), were cytogenetically characterized. Genomic gains and losses were precisely defined using microarray-based comparative genomic hybridization (array-CGH), and minimal overlapping analysis led to the identification of the common unbalanced genomic regions. Using the U133Plus 2.0 Affymetrix gene chip array, we analyzed PMR-MM7 early and late passages for genome-wide gene expression, and correlated the differentially expressed genes with copy-number changes. The presence of a high number of genetic imbalances occurring from early to late culture steps reflected the tendency of MM cells toward genomic instability. The selection of specific chromosomal abnormalities observed during subsequent cultures demonstrated the spontaneous evolution of the cancer cells in an in vitro environment. MM cell lines were characterized by copy-number changes associated with the TP53 apoptotic pathway already present at the first steps of in vitro culture. Prolonged culture led to acquisition of additional chromosomal copy-number changes associated with dysregulation of genes involved in cell adhesion, regulation of mitotic cell cycle, signal transduction, carbohydrate metabolism, motor activity, glycosaminoglycan biosynthesis, protein binding activity, lipid transport, ATP synthesis, and methyltransferase activity.


Assuntos
Aberrações Cromossômicas , Cromossomos Humanos/genética , Dosagem de Genes/genética , Perfilação da Expressão Gênica/métodos , Mesotelioma/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Mapeamento Cromossômico , Genoma Humano/genética , Humanos , Hibridização in Situ Fluorescente , Mesotelioma/metabolismo , Hibridização de Ácido Nucleico , Cariotipagem Espectral , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
17.
Int J Androl ; 30(4): 350-65, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17705808

RESUMO

The JKT-1 cell line has been used in multiple independent studies as a representative model of human testicular seminoma. However, no cell line for this specific tumour type has been independently confirmed previously; and therefore, the seminomatous origin of JKT-1 must be proven. The genetic constitution of the JKT-1 cells was determined using flow cytometry and spectral karyotyping, as well as array comparative genomic hybridization and fluorescent in situ hybridization. Marker profiling, predominantly based on differentially expressed proteins during normal germ cell development, was performed by immunohistochemistry and Western blot analyses. Moreover, genome wide affymetrix mRNA expression and profiling of 157 microRNAs was performed, and the status of genomic imprinting was determined. A germ cell origin of the JKT-1 cells was in line with genomic imprinting status and marker profile (including positive staining for several cancer-testis antigens). However, the supposed primary tumour, from which the cell line was derived, being indeed a classical seminoma, was molecularly proven not to be the origin of the cell line. The characteristic chromosomal anomalies of seminoma, e.g. gain of the short arm of chromosome 12, as well as the informative marker profile (positive staining for OCT3/4, NANOG, among others) were absent in the various JKT-1 cell lines investigated, irrespective of where the cells were cultured. All results indicate that the JKT-1 cell line is not representative of human seminoma. Although it can originate from an early germ cell, a non-germ cell derivation cannot be excluded.


Assuntos
Linhagem Celular Tumoral , Seminoma , Neoplasias Testiculares , Humanos , Imuno-Histoquímica , Cariotipagem , Masculino , Modelos Biológicos , Reprodutibilidade dos Testes
18.
Genes Chromosomes Cancer ; 45(5): 437-46, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16419055

RESUMO

Chromosome rearrangements are found in many acute leukemias. As a result, genes at the breakpoints can be disrupted, forming fusion genes. One of the genes involved in several chromosome aberrations in hematological malignancies is NUP98 (11p15). As NUP98 is close to the 11p telomere, small translocations might easily be missed. Using a NUP98-specific split-signal fluorescence in situ hybridization (FISH) probe combination, we analyzed 84 patients with acute myeloid leukemia (AML), acute lymphoblastic leukemia, or myelodysplastic syndrome with either normal karyotypes or 11p abnormalities to investigate whether there are unidentified 11p15 rearrangements. Neither NUP98 translocations nor deletions were identified in cases with normal karyotypes, indicating these aberrations may be very rare in this group. However, NUP98 deletions were observed in four cases with unbalanced 11p aberrations, indicating that the breakpoint is centromeric of NUP98. Rearrangements of NUP98 were identified in two patients, both showing 11p abnormalities in the diagnostic karyotype: a t(4;11)(q1?3;p15) with expression of the NUP98-RAP1GDS1 fusion product detected in a 60-year-old woman with AML-M0, and an add(11)(p15) with a der(21)t(11;21)(p15;p13) observed cytogenetically in a 1-year-old boy with AML-M7. JARID1A was identified as the fusion partner of NUP98 using 3' RACE, RT-PCR, and FISH. JARID1A, at 12p13, codes for retinoblastoma binding protein 2, a protein implicated in transcriptional regulation. This is the first report of JARID1A as a partner gene in leukemia.


Assuntos
Cromossomos Humanos Par 12 , Leucemia/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Doença Aguda , Adolescente , Adulto , Idoso , Sequência de Aminoácidos , Sequência de Bases , Criança , Pré-Escolar , Primers do DNA , Feminino , Humanos , Hibridização in Situ Fluorescente , Lactente , Cariotipagem , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular
19.
Genes Chromosomes Cancer ; 45(8): 731-9, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16646086

RESUMO

The t(7;12)(q36;p13) is a recurrent translocation involving the ETV6/TEL gene (12p13) and a heterogeneous breakpoint at 7q36. A fusion transcript between HLXB9 and ETV6 in AML with t(7;12) is occasionally found. To study the incidence of t(7;12) in infant and childhood acute leukemia, we screened 320 cases <36 months using FISH. Additionally, 28 pediatric cases >36 months with cytogenetic breakpoints at 12p and 7q were investigated. We studied the presence of an HXLB9-ETV6 fusion transcript and quantified the expression of various genes located in the 7q36 breakpoint region. In total, six AML patients carried the t(7;12) of which five were infants and one child of 18 months. Only one out of 99 infant ALL patients harbored the t(7;12). No t(7;12) was found in older children with AML or ALL. AML patients carrying a t(7;12) had a poor outcome with a 3-year EFS of 0%. A fusion of HLXB9 to ETV6 was found in four AML cases with t(7;12). The 7q36 genes NOM1, LMBR1, RNF32, and SHH were equally expressed among t(7;12)-positive AML versus t(7;12)-negative AML, t(7;12)-negative ALL, or normal bone marrow. However, the HLXB9 expression was highly increased in t(7;12)-positive cases, including those with an HLXB9-ETV6 fusion. We conclude that the t(7;12) is almost exclusively present in infant AML and covers 30% of infant AML, while it is extremely rare in infant ALL and older children. The t(7;12) is associated with a poor outcome and an ectopic expression of HLXB9 is commonly involved in this genetic subtype of leukemia.


Assuntos
Cromossomos Humanos Par 12 , Cromossomos Humanos Par 7 , Proteínas de Homeodomínio/genética , Leucemia Mieloide/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Fatores de Transcrição/genética , Translocação Genética , Doença Aguda , Pré-Escolar , Quebra Cromossômica , Estudos de Coortes , Feminino , Proteínas de Homeodomínio/metabolismo , Humanos , Hibridização in Situ Fluorescente , Lactente , Recém-Nascido , Masculino , Modelos Genéticos , Proteínas de Fusão Oncogênica/genética , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Repressoras/genética , Fatores de Transcrição/metabolismo , Variante 6 da Proteína do Fator de Translocação ETS
20.
Int J Cancer ; 114(6): 917-25, 2005 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-15645425

RESUMO

The BCL-2 family has been implicated in the pathogenesis of various hematopoietic malignancies, including follicular non-Hodgkin lymphoma and B-cell chronic lymphocytic leukemia. To identify genes that act synergistically in BCL2-enforced leukemogenesis, we developed a murine B-cell lymphoma/leukemia model based on the IL-3-dependent Balb/C pro-B line (FL5.12). FL5.12 cells were stably transfected with antiapoptotic BCL-2 alone or in combination with proapoptotic BAX or nonfunctional mutant BAX, thereby creating various levels of imbalance within the BCL-2 family. Transfectants were intravenously injected into normal Balb/C mice. Whereas FL5.12 cells did not provoke leukemia, mice injected with stable transfectants died of leukemia over time. Disease incidence and latency time depended on the degree of imbalance in the BCL-2 family, supporting a model whereby BCL2 drives tumorigenesis. All mice presented with hepatosplenomegaly and leukemic FL5.12 cells in peripheral blood and bone marrow compartments. Leukemic conversion was accompanied by secondary genetic aberrations leading to clonal IL-3-responsive leukemia. Cellular transformation was independent of alterations in c-Myc or downstream apoptotic pathway. Leukemic clones retained a normal DNA damage response leading to elevated P53 and P21 levels and cell cycle arrest upon irradiation. In conclusion, our mouse model may prove a valuable tool to identify genes that cooperate in BCL2-enforced lymphoma/leukemogenesis.


Assuntos
Transformação Celular Neoplásica , Leucemia de Células B/genética , Leucemia de Células B/fisiopatologia , Linfoma de Células B/genética , Linfoma de Células B/fisiopatologia , Proteínas Proto-Oncogênicas c-bcl-2/farmacologia , Animais , Dano ao DNA , Modelos Animais de Doenças , Genes p53 , Hepatomegalia , Leucemia de Células B/veterinária , Linfoma de Células B/veterinária , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Proto-Oncogênicas c-bcl-2/genética , Esplenomegalia , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA