Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34607954

RESUMO

BRCA1 germline mutations are associated with an increased risk of breast and ovarian cancer. Recent findings of others suggest that BRCA1 mutation carriers also bear an increased risk of esophageal and gastric cancer. Here, we employ a Brca1/Trp53 mouse model to show that unresolved replication stress (RS) in BRCA1 heterozygous cells drives esophageal tumorigenesis in a model of the human equivalent. This model employs 4-nitroquinoline-1-oxide (4NQO) as an RS-inducing agent. Upon drinking 4NQO-containing water, Brca1 heterozygous mice formed squamous cell carcinomas of the distal esophagus and forestomach at a much higher frequency and speed (∼90 to 120 d) than did wild-type (WT) mice, which remained largely tumor free. Their esophageal tissue, but not that of WT control mice, revealed evidence of overt RS as reflected by intracellular CHK1 phosphorylation and 53BP1 staining. These Brca1 mutant tumors also revealed higher genome mutation rates than those of control animals; the mutational signature SBS4, which is associated with tobacco-induced tumorigenesis; and a loss of Brca1 heterozygosity (LOH). This uniquely accelerated Brca1 tumor model is also relevant to human esophageal squamous cell carcinoma, an often lethal tumor.


Assuntos
Proteína BRCA1/genética , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , Perda de Heterozigosidade/genética , Proteína Supressora de Tumor p53/genética , 4-Nitroquinolina-1-Óxido/toxicidade , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Quinase 1 do Ponto de Checagem/metabolismo , Modelos Animais de Doenças , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/induzido quimicamente , Carcinoma de Células Escamosas do Esôfago/patologia , Feminino , Mutação em Linhagem Germinativa/genética , Heterozigoto , Humanos , Perda de Heterozigosidade/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
2.
Breast Cancer Res ; 22(1): 133, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33267874

RESUMO

BACKGROUND: Mutations in one allele of the TP53 gene in early stages are frequently followed by the loss of the remaining wild-type p53 (wtp53) allele (p53LOH) during tumor progression. Despite the strong notion of p53LOH as a critical step in tumor progression, its oncogenic outcomes that facilitate the selective pressure for p53LOH occurrence were not elucidated. METHODS: Using MMTV;ErbB2 mouse model of breast cancer carrying heterozygous R172H p53 mutation, we identified a novel gain-of-function (GOF) activity of mutant p53 (mutp53): the exacerbated loss of wtp53 allele in response to γ-irradiation. RESULTS: As consequences of p53LOH in mutp53 heterozygous cells, we observed profound stabilization of mutp53 protein, the loss of p21 expression, the abrogation of G2/M checkpoint, chromosomal instability, centrosome amplification, and transcriptional upregulation of mitotic kinase Nek2 (a member of Never in Mitosis (NIMA) Kinases family) involved in the regulation of centrosome function. To avoid the mitotic catastrophe in the absence of G2/M checkpoint, cells with centrosome amplification adapt Nek2-mediated centrosomes clustering as pro-survival mutp53 GOF mechanism enabling unrestricted proliferation and clonal expansion of cells with p53LOH. Thus, the clonal dominance of mutp53 cells with p53LOH may represent the mechanism of irradiation-induced p53LOH. We show that pharmacological and genetic ablation of Nek2 decreases centrosome clustering and viability of specifically mutp53 cells with p53LOH. CONCLUSION: In a heterogeneous tumor population, Nek2 inhibition may alter the selective pressure for p53LOH by contraction of the mutp53 population with p53LOH, thus, preventing the outgrowth of genetically unstable, more aggressive cells.


Assuntos
Neoplasias da Mama/genética , Perda de Heterozigosidade/genética , Quinases Relacionadas a NIMA/genética , Proteína Supressora de Tumor p53/genética , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Centrossomo/metabolismo , Instabilidade Cromossômica , Conjuntos de Dados como Assunto , Modelos Animais de Doenças , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Mutação com Ganho de Função , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Perda de Heterozigosidade/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Quinases Relacionadas a NIMA/antagonistas & inibidores , Receptor ErbB-2/genética , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
3.
Eur J Cancer ; 112: 66-79, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30947024

RESUMO

BACKGROUND: Induction chemotherapy plays an important role in the management of patients with high-risk neuroblastoma. Predictors of response to induction therapy are largely lacking. We sought to describe clinical and biological features associated with induction response. METHODS: Patients from four consecutive COG high-risk trials were included. Response was evaluated by the 1993 International Neuroblastoma Response Criteria. The primary end-point was end-induction partial response (PR) or better. Univariate analyses were performed to compare response as a function of clinical or biologic predictors. A multivariate logistic regression model using significant predictors from univariate analyses was constructed to model PR or better. RESULTS: The analytic cohort included 1242 patients. End-induction response ≥PR was significantly associated with higher event-free and overall survival. Baseline factors associated with ≥PR included age <18 months (87.4% with ≥PR vs. 78.7% if older; p = 0.0103), International Neuroblastoma Staging System non-stage 4 (89.0% vs. 78.4% if stage 4; p = 0.0016), MYCN amplification (85.5% vs. 77.1% if non-amplified; p = 0.0006), 1p loss of heterozygosity (LOH; 85.6% vs. 76.0% if no LOH; p = 0.0085), no 11q LOH (84.8% vs. 70.9% if 11q LOH; p = 0.0004) and high mitosis-karyorrhexis index (MKI; 84.5% vs. 77.5% if low-intermediate MKI; p = 0.0098). On multivariable analysis (n = 407), the absence of 11q LOH was the only factor that remained significantly associated with ≥PR (odds ratio: 1.962 vs. 11q LOH; 95% confidence interval 1.104-3.487; p = 0.0216). CONCLUSIONS: Improved end-induction response in high-risk neuroblastoma is associated with longer survival. Patients with 11q LOH are less likely to respond to induction therapies and should be prioritised for novel approaches in future trials.


Assuntos
Neuroblastoma/tratamento farmacológico , Pré-Escolar , Estudos de Coortes , Intervalo Livre de Doença , Feminino , Amplificação de Genes/efeitos dos fármacos , Amplificação de Genes/genética , Humanos , Lactente , Modelos Logísticos , Perda de Heterozigosidade/efeitos dos fármacos , Perda de Heterozigosidade/genética , Masculino , Proteína Proto-Oncogênica N-Myc/genética , Terapia Neoadjuvante/métodos , Estadiamento de Neoplasias/métodos , Neuroblastoma/genética , Neuroblastoma/patologia , Proteínas Nucleares/genética , Prognóstico , Fatores de Risco
4.
Drug Chem Toxicol ; 42(4): 394-402, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29681187

RESUMO

Bendamustine, an anticancer drug with alkylating properties, is widely used to treat hematological malignancies. Since the nitrogen mustard family alkylators induce DNA damages and have been associated with an elevated risk of second malignancy, current study evaluates the cytotoxic, mutagenic, and recombinogenic effects of bendamustine by using, respectively the mitotic index assay, the in vitro mammalian cell micronucleus test (Mnvit) and the chromosome aberration (CA) test in human peripheral lymphocytes, and the in vivo homozygotization assay in Aspergillus nidulans, which detects the loss of heterozygosity (LOH) due to somatic recombination. Bendamustine (6.0 µg/ml, 9.0 µg/ml, and 12.0 µg/ml) induced a statistically significant concentration-related increase in the frequencies of micronuclei and a significant reduction in the cytokinesis block proliferation index (CBPI) rates when compared to negative control. In the CA test, bendamustine significantly increased the frequencies of structural aberrations at the three tested concentrations when compared to the negative control. Aspergillus nidulans diploids, obtained after bendamustine treatment (6.0 µg/ml, 12.0 µg/ml, and 24.0 µg/ml), produced, after haploidization, homozygotization index (HI) rates higher than 2.0 and significantly different from the negative control. Since bendamustine showed genotoxic effects in all tested concentrations, two of them corresponding to the peak plasma concentrations observed in cancer patients treated with bendamustine, data provided in the current research work may be useful to identify the most appropriate dosage regimen to achieve the efficacy and safety of this anticancer medication.


Assuntos
Antineoplásicos Alquilantes/toxicidade , Aspergillus nidulans/efeitos dos fármacos , Cloridrato de Bendamustina/toxicidade , Aberrações Cromossômicas/induzido quimicamente , Perda de Heterozigosidade/efeitos dos fármacos , Linfócitos/efeitos dos fármacos , Adolescente , Adulto , Aspergillus nidulans/genética , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Linfócitos/patologia , Micronúcleos com Defeito Cromossômico/induzido quimicamente , Testes para Micronúcleos , Adulto Jovem
5.
Genetics ; 203(3): 1161-76, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27206717

RESUMO

By testing the susceptibility to DNA damaging agents of several Candida albicans mutant strains derived from the commonly used laboratory strain, CAI4, we uncovered sensitivity to methyl methanesulfonate (MMS) in CAI4 and its derivatives, but not in CAF2-1. This sensitivity is not a result of URA3 disruption because the phenotype was not restored after URA3 reintroduction. Rather, we found that homozygosis of a short region of chromosome 3R (Chr3R), which is naturally heterozygous in the MMS-resistant-related strains CAF4-2 and CAF2-1, confers MMS sensitivity and modulates growth polarization in response to MMS. Furthermore, induction of homozygosity in this region in CAF2-1 or CAF4-2 resulted in MMS sensitivity. We identified 11 genes by SNP/comparative genomic hybridization containing only the a alleles in all the MMS-sensitive strains. Four candidate genes, SNF5, POL1, orf19.5854.1, and MBP1, were analyzed by generating hemizygous configurations in CAF2-1 and CAF4-2 for each allele of all four genes. Only hemizygous MBP1a/mbp1b::SAT1-FLIP strains became MMS sensitive, indicating that MBP1a in the homo- or hemizygosis state was sufficient to account for the MMS-sensitive phenotype. In yeast, Mbp1 regulates G1/S genes involved in DNA repair. A second region of homozygosis on Chr2L increased MMS sensitivity in CAI4 (Chr3R homozygous) but not CAF4-2 (Chr3R heterozygous). This is the first example of sign epistasis in C. albicans.


Assuntos
Candida albicans/genética , Epistasia Genética , Proteínas Fúngicas/genética , Perda de Heterozigosidade/genética , Alelos , Antifúngicos/toxicidade , Candida albicans/efeitos dos fármacos , Hibridização Genômica Comparativa , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Proteínas Fúngicas/biossíntese , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Perda de Heterozigosidade/efeitos dos fármacos , Metanossulfonato de Metila/toxicidade , Polimorfismo de Nucleotídeo Único
6.
Tsitol Genet ; 50(5): 17-24, 2016.
Artigo em Inglês, Russo | MEDLINE | ID: mdl-30480912

RESUMO

Precise chromosome segregation is vital for speciation and hybrid formation. The aim of the work was to study the chromosomes behaviour and inheritance of maternal and paternal genomes in Arabidopsis regenerants de-rived from in vitro cultured cells on the medium with PFFA. The Arabidopsis thaliana model hybrid between Columbia and Landsberg erecta ecotypes was developed, which chromosomes were easy to distinguish using the 12 SSLP selected markers. Also, the influence of PFFA on the callus formation and regeneration of plants was analysed. 20 regenerated plants cultured with PFFA were derived, three of which were shown to loss the heterozygosity in six loci by DNA markers analysis. Different models are certainly required to understand how and when the mechanisms leading to proper chromosome segregation are established in species and hybrids.


Assuntos
Arabidopsis/efeitos dos fármacos , Cromossomos de Plantas/efeitos dos fármacos , DNA de Plantas/genética , Perda de Heterozigosidade/efeitos dos fármacos , p-Fluorfenilalanina/farmacologia , Arabidopsis/genética , Arabidopsis/metabolismo , Quimera , Mapeamento Cromossômico , Segregação de Cromossomos , Cruzamentos Genéticos , DNA de Plantas/metabolismo , Loci Gênicos , Marcadores Genéticos
7.
PLoS One ; 10(8): e0134426, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26247786

RESUMO

BACKGROUND: The majority of chordomas show activation of the platelet-derived growth factor receptor (PDGFR). Based on in vitro intertumoral variation in response to recombinant PDGF protein and PDGFR inhibition, and variable tumor response to imatinib, we hypothesized that chordomas resistant to PDGFR inhibition may possess downstream activation of the pathway. METHODS: Molecular profiling was performed on 23 consecutive chordoma primary tissue specimens. Primary cultures established from 20 of the 23 specimens, and chordoma cell lines, UCH-1 and UCH-2, were used for in vitro experiments. RESULTS: Loss of heterozygosity (LOH) at the phosphatase and tensin homolog (PTEN) locus was observed in 6 specimens (26%). PTEN disruption statistically correlated with increased Ki-67 proliferation index, an established marker of poor outcome for chordoma. Compared to wild type, PTEN deficient chordomas displayed increased proliferative rate, and responded less favorably to PDGFR inhibition. PTEN gene restoration abrogated this growth advantage. Chordomas are characterized by intratumoral hypoxia and local invasion, and histone deacetylase (HDAC) inhibitors are capable of attenuating both hypoxic signaling and cell migration. The combination of PDGFR and HDAC inhibition effectively disrupted growth and invasion of PTEN deficient chordoma cells. CONCLUSIONS: Loss of heterozygosity of the PTEN gene seen in a subset of chordomas is associated with aggressive in vitro behavior and strongly correlates with increased Ki-67 proliferative index. Combined inhibition of PDGFR and HDAC attenuates proliferation and invasion in chordoma cells deficient for PTEN.


Assuntos
Inibidores de Histona Desacetilases/farmacologia , Perda de Heterozigosidade/efeitos dos fármacos , PTEN Fosfo-Hidrolase/genética , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Adulto , Idoso , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Criança , Cordoma/metabolismo , Cordoma/patologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Masculino , Repetições de Microssatélites/genética , Pessoa de Meia-Idade , PTEN Fosfo-Hidrolase/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/análise , Adulto Jovem
8.
Food Chem Toxicol ; 84: 55-63, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26190540

RESUMO

In this study, we evaluated the toxic and genotoxic potential of zinc oxide nanoparticles (ZnO NPs) of 20 nm and the mutagenic potential of these ZnO NPs as well as that of an amorphous ZnO. Toxicity was assessed by XTT colorimetric assay. ZnO NPs were toxic at concentrations equal to or higher than 240.0 µM. Genotoxicity was assessed by in vitro Cytokinesis Block Micronucleus Assay (CBMN) in V79 cells. ZnO NPs were genotoxic at 120.0 µM. The mutagenic potential of amorphous ZnO and the ZnO NPs was assayed using the wing Somatic Mutation and Recombination Test (SMART) of Drosophila melanogaster. In the Standard cross, the amorphous ZnO and ZnO NPs were not mutagenic. Nevertheless, Marker trans-heterozygous individuals from the High bioactivation cross treated with amorphous ZnO (6.25 mM) and ZnO NPs (12.50 mM) displayed a significant increased number of mutant spots when compared with the negative control. In conclusion, the results were not dose related and indicate that only higher concentrations of ZnO NPs were toxic and able to induce genotoxicity in V79 cells. The increase in mutant spots observed in D. melanogaster was generated due to mitotic recombination, rather than mutational events.


Assuntos
Nanopartículas Metálicas/toxicidade , Mutagênese/efeitos dos fármacos , Mutagênicos/toxicidade , Recombinação Genética/efeitos dos fármacos , Óxido de Zinco/toxicidade , Animais , Animais Geneticamente Modificados , Bioensaio , Linhagem Celular , Cricetulus , Cruzamentos Genéticos , Drosophila melanogaster , Feminino , Marcadores Genéticos/efeitos dos fármacos , Perda de Heterozigosidade/efeitos dos fármacos , Masculino , Nanopartículas Metálicas/ultraestrutura , Testes para Micronúcleos , Testes de Mutagenicidade , Mutagênicos/química , Pigmentação/efeitos dos fármacos , Asas de Animais , Óxido de Zinco/química
9.
Nature ; 518(7538): 240-4, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25409150

RESUMO

Broad and deep tumour genome sequencing has shed new light on tumour heterogeneity and provided important insights into the evolution of metastases arising from different clones. There is an additional layer of complexity, in that tumour evolution may be influenced by selective pressure provided by therapy, in a similar fashion to that occurring in infectious diseases. Here we studied tumour genomic evolution in a patient (index patient) with metastatic breast cancer bearing an activating PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha, PI(3)Kα) mutation. The patient was treated with the PI(3)Kα inhibitor BYL719, which achieved a lasting clinical response, but the patient eventually became resistant to this drug (emergence of lung metastases) and died shortly thereafter. A rapid autopsy was performed and material from a total of 14 metastatic sites was collected and sequenced. All metastatic lesions, when compared to the pre-treatment tumour, had a copy loss of PTEN (phosphatase and tensin homolog) and those lesions that became refractory to BYL719 had additional and different PTEN genetic alterations, resulting in the loss of PTEN expression. To put these results in context, we examined six other patients also treated with BYL719. Acquired bi-allelic loss of PTEN was found in one of these patients, whereas in two others PIK3CA mutations present in the primary tumour were no longer detected at the time of progression. To characterize our findings functionally, we examined the effects of PTEN knockdown in several preclinical models (both in cell lines intrinsically sensitive to BYL719 and in PTEN-null xenografts derived from our index patient), which we found resulted in resistance to BYL719, whereas simultaneous PI(3)K p110ß blockade reverted this resistance phenotype. We conclude that parallel genetic evolution of separate metastatic sites with different PTEN genomic alterations leads to a convergent PTEN-null phenotype resistant to PI(3)Kα inhibition.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Resistencia a Medicamentos Antineoplásicos/genética , PTEN Fosfo-Hidrolase/deficiência , PTEN Fosfo-Hidrolase/genética , Inibidores de Fosfoinositídeo-3 Quinase , Tiazóis/farmacologia , Alelos , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Classe I de Fosfatidilinositol 3-Quinases , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Perda de Heterozigosidade/efeitos dos fármacos , Perda de Heterozigosidade/genética , Camundongos , Camundongos Nus , PTEN Fosfo-Hidrolase/metabolismo , Tiazóis/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Ecotoxicology ; 22(5): 889-99, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23604582

RESUMO

Microevolution due to pollution can occur mainly through genetic drift bottlenecks, especially of small sized populations facing intense lethal pulses of contaminants, through mutations, increasing allelic diversity, and through natural selection, with the disappearance of the most sensitive genotypes. This loss of genotypes can lead to serious effects if coupled to specific hypothetical scenarios. These may be categorized as leading, first, to the loss of alleles-the recessive tolerance inheritance hypothesis. Second, leading to a reduction of the population growth rate-the mutational load and fitness costs hypotheses. Third, leading to an increased susceptibility of further genetic erosion both at future inputs of the same contaminant-differential physiological recovery, endpoints (dis)association, and differential phenotypic plasticity hypotheses-and at sequential or simultaneous inputs of other contaminants-the multiple stressors differential tolerance hypothesis. Species in narrowly fluctuating environments (tropics and deep sea) may have a particularly high susceptibility to genetic erosion-the Plus ça change (plus c'est la meme chose) hypothesis. A discussion on the consequences of these hypotheses is what this essay aimed at.


Assuntos
Adaptação Fisiológica/efeitos dos fármacos , Poluentes Ambientais/toxicidade , Deriva Genética , Aptidão Genética/efeitos dos fármacos , Variação Genética , Perda de Heterozigosidade/efeitos dos fármacos , Adaptação Fisiológica/genética , Ecossistema , Evolução Molecular , Interação Gene-Ambiente , Genética Populacional , Mutação
11.
J Appl Toxicol ; 33(3): 209-13, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22015851

RESUMO

Benign prostatic hyperplasia (BPH) is the most common tumor in men over 40 years of age. Acute urinary retention (AUR) is regarded as the most serious hazard of untreated BPH. α-Blockers, such as doxazosin mesylate, and 5-α reductase inhibitors, such as finasteride, are frequently used because they decrease both AUR and the need for BPH-related surgery. An extract of the fruit from American saw palmetto plant has also been used as an alternative treatment for BPH. The paucity of information available concerning the genotoxic action of these compounds led us to assess their activity as inducers of different types of DNA lesions using the somatic mutation and recombination test in Drosophila melanogaster. Finasteride did not induce gene mutation, chromosomal mutation or mitotic recombination, which means it was nongenotoxic in our experimental conditions. On the other hand, doxazosin mesylate and saw palmetto induced significant increases in spot frequencies in trans-heterozygous flies. In order to establish the actual role played by mitotic recombination and by mutation in the genotoxicity observed, the balancer-heterozygous flies were also analyzed, showing no increment in the total spot frequencies in relation to the negative control, for both drugs. Doxazosin mesylate and saw palmetto were classified as specific inducers of homologous recombination in Drosophila proliferative cells, an event linked to the loss of heterozygosity.


Assuntos
Anti-Hipertensivos/toxicidade , Doxazossina/toxicidade , Drosophila/efeitos dos fármacos , Mutagênicos/toxicidade , Extratos Vegetais/toxicidade , Recombinação Genética/efeitos dos fármacos , Animais , DNA/efeitos dos fármacos , Dano ao DNA , Drosophila/genética , Feminino , Perda de Heterozigosidade/efeitos dos fármacos , Testes de Mutagenicidade , Serenoa , Asas de Animais/efeitos dos fármacos , Asas de Animais/crescimento & desenvolvimento
12.
PLoS One ; 7(10): e47012, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23056560

RESUMO

WNT7A (wingless-type MMTV integration site family, member 7A) is a known tumor suppressor gene of non-small cell lung carcinomas (NSCLC) and is frequently inactivated due to CpG-island hypermethylation in human cancers. The members of WNT family are involved in cell signaling and play crucial roles in cancer development. In the present work hypermethylation of the WNT7A gene was detected in 66% (29/44) of analyzed clear cell renal cell carcinomas (RCCs) using methyl-specific PCR (MSP). Moreover, bisulfite sequencing confirmed intensive hypermethylation of the 5'-CpG island of the WNT7A gene. Methylation analysis revealed positive correlations between tumor stage, Fuhrman nuclear grade and WNT7A hypermethylation. Additionally, restoration of WNT7A gene expression in the A498 cell line by 5-aza-2'-deoxycytidine treatment confirmed a direct contribution of hypermethylation in silencing of the WNT7A gene. High frequency of loss of heterozygosity (LOH) was demonstrated on chromosome 3p25 in regions surrounding the WNT7A gene. The frequent down-regulation of WNT7A gene expression was detected in 88% (15/17) of clear cell RCCs. We have also shown that the WNT7A gene possesses tumor suppression function by colony-formation and cell proliferation assays in RCC cell lines. In summary, the WNT7A gene is inactivated by genetic/epigenetic alterations in clear cell RCC and demonstrates tumor suppressor properties.


Assuntos
Carcinoma de Células Renais/genética , Proteínas Wnt/genética , Adulto , Idoso , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Decitabina , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/genética , Feminino , Marcadores Genéticos/genética , Humanos , Perda de Heterozigosidade/efeitos dos fármacos , Perda de Heterozigosidade/genética , Masculino , Repetições de Microssatélites/genética , Pessoa de Meia-Idade , Adulto Jovem
13.
Mutagenesis ; 26(6): 771-81, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21775299

RESUMO

There is some evidence that the mouse lymphoma TK assay (MLA) can detect aneugens, and this is accepted in the current International Conference on Harmonisation guidance for testing pharmaceuticals. However, whether or not it can be used as a reliable screen for aneugenicity has been the subject of debate. Consequently, aneugens with diverse mechanisms of action were tested in the MLA using 24-h exposure. No evidence of increased mutant frequency was seen with noscapine, diazepam or colchicine and increases were seen with taxol, carbendazim, econazole and chloral hydrate only at high levels of toxicity (for all but one taxol concentration survival reduced to ≤10% of control). None of these agents would be unequivocally classified as positive using currently accepted criteria. The largest increases in mutant number were seen with taxol and carbendazim; therefore, trifluorothymidine (TFT)-resistant clones resulting from treatment with them were cultured and analysed for chromosome 11 copy number using fluorescent in situ hybridisation (FISH) and loss of heterozygosity (LOH). High concentrations of these aneugens induced LOH at all loci examined indicating only one chromosome 11 was present but, perhaps surprisingly, all were found to have two copies of chromosome 11 using FISH. This would be consistent with loss of the tk(+) chromosome 11b with concomitant duplication of chromosome 11a, which has been proposed as a likely mechanism for induction of TFT-resistant clones. However, it was also surprising that analysis of centromere size showed that almost all the clones had both small and large centromeres, i.e. suggesting the presence of both chromosomes 11a and 11b. In conclusion, it appears that the TFT-resistant mutants resulting from treatment with toxic concentrations of some aneugens such as taxol and carbendazim have undergone complex genetic changes. However, these data show that the MLA cannot be used as a routine screen to detect aneugens.


Assuntos
Aneugênicos/análise , Ensaios Enzimáticos/métodos , Linfoma/metabolismo , Timidina Quinase/metabolismo , Aneugênicos/toxicidade , Animais , Linhagem Celular Tumoral , Centrômero/efeitos dos fármacos , Centrômero/metabolismo , Cromossomos de Mamíferos/genética , Dosagem de Genes/efeitos dos fármacos , Dosagem de Genes/genética , Hibridização in Situ Fluorescente , Cariotipagem , Perda de Heterozigosidade/efeitos dos fármacos , Perda de Heterozigosidade/genética , Camundongos , Repetições de Microssatélites/genética , Reação em Cadeia da Polimerase
14.
Mutat Res ; 718(1-2): 33-7, 2011 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-21073980

RESUMO

In view of the scarcely available information on the in vivo mutagenic and co-mutagenic activity of nickel, the genotoxic potential of two nickel-compounds, nickel chloride (NiCl(2)) and nickel sulphate (NiSO(4)), was assessed in Drosophila melanogaster by measuring two different genetic endpoints. On the one hand, we used the wing-spot assay, which is based on the principle that the loss of heterozygosity of two suitable recessive markers, multiple wing hairs (mwh) and flare-3 (flr(3)), can lead to the formation of mutant clones in the imaginal disks of larval cells. On the other hand, the in vivo comet assay, which detects single- and double-strand DNA breaks, was also used with larval haemocytes. These cells offer several advantages: they are highly sensitive to genotoxic agents, the sampling and processing methodologies are quite simple and the level of basal DNA damage is relatively low. No significant increases in the frequencies of the three categories of mutant spots (i.e. small single spots, large single spots, and twin spots) were observed in the wing-spot assay; however, NiSO(4) induced significant dose-dependent increases in DNA damage in the comet assay. In addition, the combined treatments with gamma-radiation and NiCl(2) and NiSO(4) showed a slight but significant increase in the frequency of the three categories of mutant spots compared with the frequency induced by gamma-radiation alone, indicating that both nickel compounds have a synergistic interaction. These results support the assumption that both nickel compounds could act as co-mutagens interfering with DNA-repair processes and that the in vivo comet assay is a sensitive and effective method for detecting the DNA damage induced by NiSO(4) in haemocytes of D. melanogaster.


Assuntos
Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/genética , Mutagênicos/toxicidade , Níquel/toxicidade , Animais , Ensaio Cometa , Dano ao DNA , Proteínas de Drosophila/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/efeitos da radiação , Sinergismo Farmacológico , Feminino , Raios gama/efeitos adversos , Genes de Insetos/efeitos dos fármacos , Hemócitos/efeitos dos fármacos , Hemócitos/metabolismo , Hemócitos/efeitos da radiação , Perda de Heterozigosidade/efeitos dos fármacos , Masculino , Testes de Mutagenicidade , Mutagênicos/administração & dosagem , Níquel/administração & dosagem , Asas de Animais/efeitos dos fármacos , Asas de Animais/crescimento & desenvolvimento , Asas de Animais/efeitos da radiação
15.
Clin Cancer Res ; 16(18): 4553-60, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20823145

RESUMO

Hypoxia exists in every solid tumor and is associated with poor prognosis because of both local and systemic therapeutic resistance. Recent studies have focused on the interaction between tumor cell genetics and the dynamic state of oxygenation and metabolism. Hypoxia generates aggressive tumor cell phenotypes in part owing to ongoing genetic instability and a "mutator" phenotype. The latter may be due to suppression of DNA mismatch repair (MMR), nucleotide excision repair (NER), and double-strand break (DSB) repair. We propose a theoretical model in which hypoxia-mediated defects in DNA repair can lead to "contextual loss of heterozygosity" and drive oncogenesis. Additionally, hypoxia-mediated repair defects can be specifically targeted by DNA damaging agents and/or "contextual synthetic lethality" to kill repair-deficient cells and preserve the therapeutic ratio. These proposed concepts support the interrogation of solid tumors to document repair defects in both oxic and hypoxic tumor subregions as a conduit to novel clinical trials within the context of personalized medicine.


Assuntos
Carcinógenos/toxicidade , Reparo do DNA/genética , Perda de Heterozigosidade/fisiologia , Neoplasias/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/fisiologia , Carcinógenos/farmacologia , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/fisiologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/fisiologia , Instabilidade Genômica/efeitos dos fármacos , Instabilidade Genômica/fisiologia , Humanos , Perda de Heterozigosidade/efeitos dos fármacos , Perda de Heterozigosidade/genética , Modelos Biológicos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/radioterapia , Falha de Tratamento
16.
Br J Cancer ; 103(3): 340-6, 2010 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-20606684

RESUMO

BACKGROUND: Colorectal cancer is (CRC) one of the commonest cancers and its therapy is still based on few drugs. Currently, no biological criteria are used to choose the most effective of the established drugs for treatment. METHODS: A panel of 77 CRC cell lines was tested for sensitivity to 5-fluorouracil (5FU) using the SRB assay. The responses were grouped into three categories and correlated with genetic changes in the cell lines. RESULTS: The strongest and most clearcut correlation was between 5-fluorouracil response and replication error status (mismatch repair deficiency). All the other significant correlations (loss of heterozygosity for DCC and mutations in TGFbIIR) are secondary to the association with replication error status. INTERPRETATION AND CONCLUSION: Our findings validate previous analyses based mainly on clinical data, and indicate that replication error status could be a useful guide to 5-fluorouracil-based CRC therapy. Essentially, all previously described correlations with 5FU response are secondary to the association with replication error status.


Assuntos
Linhagem Celular Tumoral/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Reparo de Erro de Pareamento de DNA/efeitos dos fármacos , Fluoruracila/toxicidade , Antimetabólitos Antineoplásicos/toxicidade , Materiais Biocompatíveis , Colágeno , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Replicação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Humanos , Laminina , Perda de Heterozigosidade/efeitos dos fármacos , Mutação , Proteoglicanas
17.
Mutat Res ; 696(2): 139-43, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20083227

RESUMO

Recent studies have added paclitaxel (PAC) to traditional cisplatin (CIS) regimen to treat squamous cell carcinoma of the head and neck. The target of these antineoplastic agents is nuclear DNA for CIS and microtubules for PAC, although it is not restricted to malignant cells. In this study, the genotoxicity of the combined treatment of PAC and CIS was investigated using the standard version of the wing Somatic Mutation and Recombination Test (SMART) in Drosophila melanogaster. Quantitative and qualitative genotoxic effects of these compounds were estimated by comparing wing spot frequencies in marker-heterozygous to balancer-heterozygous flies. Two different concentrations of PAC (0.0025 and 0.005mM) and CIS (0.025 and 0.05mM) as well as combinations of them were employed. The results demonstrated that the spindle poison PAC alone was not genotoxic in this test system, while CIS was able to induce a high incidence of DNA damage in both genotypes, mainly related to somatic recombination. The data obtained for the combined treatments showed that its genotoxicity varied with the concentrations used. In small concentrations the number of total spots induced by combination was reduced in relation to CIS 0.025mM just for marker-heterozygous flies, showing that somatic recombination was the prevalent event involved. At higher concentrations the combined treatment showed significant reductions in the frequencies of large single spots, for both genotypes, and twin spots for marker-heterozygous flies, but did not significantly reduce the total spots frequency in either genotype. The data suggest that aneugenic activity of PAC could be responsible for the reduction in the genotoxicity of CIS.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Cisplatino/toxicidade , Drosophila melanogaster/efeitos dos fármacos , Paclitaxel/toxicidade , Animais , Cisplatino/administração & dosagem , Dano ao DNA/efeitos dos fármacos , Perda de Heterozigosidade/efeitos dos fármacos , Testes de Mutagenicidade , Mutagênicos , Paclitaxel/administração & dosagem
18.
Drug Chem Toxicol ; 32(4): 338-43, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19793026

RESUMO

The recombinogenic potential of fluoxetine, an antidepressant widely prescribed in the treatment of depressive disorders in cancer patients, was investigated in this study. A heterozygous diploid strain of Aspergillus nidulans was utilized. Fluoxetine at 7.5, 15, and 30 microM concentrations induced homozygosity of several nutritional genetic markers and significantly increased their homozygotization index values. Since mitotic recombination is a mechanism leading to malignant growth through the loss of a functional copy of a heterozygous tumor-suppressor gene, fluoxetine may be characterized as an inducer of secondary malignancies in cancer patients after antidepressant treatment.


Assuntos
Antidepressivos de Segunda Geração/farmacologia , Aspergillus nidulans/efeitos dos fármacos , Fluoxetina/farmacologia , Perda de Heterozigosidade/efeitos dos fármacos , Mutação/efeitos dos fármacos , Recombinação Genética/efeitos dos fármacos , Antidepressivos de Segunda Geração/efeitos adversos , Aspergillus nidulans/genética , Troca Genética , Diploide , Relação Dose-Resposta a Droga , Fluoxetina/efeitos adversos , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Heterozigoto , Humanos , Perda de Heterozigosidade/genética , Testes de Sensibilidade Microbiana , Mutação/fisiologia
19.
Asian Pac J Cancer Prev ; 10(4): 715-6, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19827902

RESUMO

Chewing Khaini damages chromosomes, in the form of loss of heterozygosity (LOH), identified on the long arm of chromosome 2 (2q), the short arm of chromosome 3 (3p) and the long arm of chromosome 21 (21q) of oral cancer cases who had quid chewing habit of more than 10 years duration, and chewed 10-15 times a day.


Assuntos
Aberrações Cromossômicas/efeitos dos fármacos , Cromossomos Humanos Par 21 , Cromossomos Humanos Par 2 , Cromossomos Humanos Par 3 , Perda de Heterozigosidade/efeitos dos fármacos , Tabaco sem Fumaça/efeitos adversos , Ásia , Humanos
20.
Int J Oncol ; 35(5): 1061-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19787260

RESUMO

Cancer progression has been associated with an increase in genomic instability indicated by inactivation of tumor suppressor genes and activation of oncogenes. Epidemiological and experimental evidence has implicated estrogens in the etiology of breast cancer. To study environmental organophosphorous pesticides is of interest since evidence indicate that pesticides may enhance cell division, increasing the risk of breast cancer. The aim was to evaluate the effects of these pesticides, such as parathion and malathion in the presence of estrogen on malignant transformation as well as on genomic instability, that is in the frequency of loss of heterozygosity (LOH) and microsatellite instability (MSI). The MCF-10F immortalized human breast epithelial cell line, that was treated with parathion or malathion alone and in combination with estrogen was used. These studies indicated that either pesticide alone or in combination with estrogen induced malignant transformation as shown by anchorage-independent growth capability and invasive characteristics in comparison to control. Such malignant phenotypic characteristics were corroborated by significant (P<0.05) increase in p53 and c-Ha-ras protein expression. Results indicated different degrees of allelic imbalance in the form of LOH or MSI with different microsatellite markers. MSI was found in malathion and estrogen-treated cells with a marker used for p53 tumor suppressor gene at loci 17p13.1. The same combination of substances presented MSI with a marker used for c-Ha-ras mapped in chromosome 11p14.1, as well as mutations in c-Ha-ras for codons 12 and 61. LOH was observed in codon 12 in the presence of estrogen or malathion alone. Parathion alone and combined with estrogen induced MSI in codon 61. It can be concluded that the organophosphorous pesticides parathion and malathion induced malignant transformation of breast cells through genomic instability altering p53 and c-Ha-ras, considered pivotal to cancer process.


Assuntos
Neoplasias da Mama/genética , Estrogênios/toxicidade , Genes p53/efeitos dos fármacos , Genes ras/efeitos dos fármacos , Compostos Organofosforados/toxicidade , Praguicidas/toxicidade , Western Blotting , Linhagem Celular Transformada , Transformação Celular Neoplásica/induzido quimicamente , Análise Mutacional de DNA , Células Epiteliais/efeitos dos fármacos , Feminino , Humanos , Perda de Heterozigosidade/efeitos dos fármacos , Malation/toxicidade , Instabilidade de Microssatélites/efeitos dos fármacos , Paration/toxicidade , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...