Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biol Proced Online ; 20: 19, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30337841

RESUMO

BACKGROUND: Personalised medicine is nowadays a major objective in oncology. Molecular characterization of tumours through NGS offers the possibility to find possible therapeutic targets in a time- and cost-effective way. However, the low quality and complexity of FFPE DNA samples bring a series of disadvantages for massive parallel sequencing techniques compared to high-quality DNA samples (from blood cells, cell cultures, etc.). RESULTS: We performed several experiments to understand the behaviour of FFPE DNA samples during the construction of SureSelectQXT libraries. First, we designed a quality checkpoint for FFPE DNA samples based on the quantification of their amplification capability (qcPCR). We observed that FFPE DNA samples can be classified according to DIN value and qcPCR concentration into unusable, or low-quality (LQ) and good-quality (GQ) DNA. For GQ samples, we increased the amount of input DNA to 150 ng and the digestion time to 30 min, whereas for LQ samples, we used 50 ng of DNA as input but we decreased the digestion time to 1 min. In all cases, we increased the cycles of the pre-hyb PCR to 10 but decreased the cycles of the post-hyb PCR to 8. In addition, we confirmed that using half of the volume of reagents can be beneficial. Finally, in order to obtain better results, we designed a decision flow-chart to achieve a seeding concentration of 12-14 pM for MiSeq Reagent Kit v2. CONCLUSIONS: Our experiments allowed us to unveil the behaviour of low-quality FFPE DNA samples during the construction of SureSelectQXT libraries. Sequencing results showed that, using our modified SureSelectQXT protocol, the final percentage of usable reads for low-quality samples was increased more than three times allowing to reach median depth/million reads values of 76.35. This value is equivalent to ~ 0.9 and ~ 0.7 of the values obtained for good-quality FFPE and high-quality DNA respectively.

2.
Pharmacogenomics J ; 11(2): 121-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20212519

RESUMO

Neurotoxicity is one of the most relevant dose-limiting toxicities of the anticancer drug paclitaxel. It exhibits substantial interindividual variability of unknown molecular basis, and represents one of the major challenges for the improvement of paclitaxel therapy. The extensive variability in paclitaxel clearance and metabolism lead us to investigate the association between polymorphisms in paclitaxel elimination pathway and neurotoxicity. We selected 13 relevant polymorphisms in genes encoding paclitaxel metabolizing enzymes (CYP2C8, CYP3A4 and CYP3A5) and transporters (organic anion transporting polypeptide (OATP) 1B1, OATP1B3 and P-glycoprotein) and genotyped them in 118 Spanish cancer patients treated with paclitaxel. After adjusting for age and treatment schedule, CYP2C8 Haplotype C and CYP3A5*3 were associated with protection (hazard ratio (HR) (per allele)=0.55; 95% confidence interval (CI)=0.34-0.89; P=0.014 and HR (per allele)=0.51; 95%CI=0.30-0.86; and P=0.012, respectively) and CYP2C8*3 with increased risk (HR (per allele)=1.72; 95%CI=1.05-2.82; and P=0.032). In each case, the allele causing increased paclitaxel metabolism was associated with increased neurotoxicity, suggesting an important role for metabolism and hydroxylated paclitaxel metabolites. We estimated the HR per paclitaxel-metabolism increasing allele carried across the three polymorphisms to be HR=1.64 (95% CI=1.26-2.14; P=0.0003). The results for P-glycoprotein were inconclusive, and no associations were observed for the other genes studied. The incorporation of this genetic data in treatment selection could help to reduce neurotoxicity events, thereby individualizing paclitaxel pharmacotherapy. These results warrant validation in independent series.


Assuntos
Antineoplásicos Fitogênicos/efeitos adversos , Hidrocarboneto de Aril Hidroxilases/genética , Citocromo P-450 CYP3A/genética , Neoplasias/tratamento farmacológico , Síndromes Neurotóxicas/etiologia , Paclitaxel/efeitos adversos , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Idoso , Alelos , Antineoplásicos Fitogênicos/uso terapêutico , Citocromo P-450 CYP2C8 , Feminino , Estudos de Associação Genética , Haplótipos , Humanos , Masculino , Pessoa de Meia-Idade , Síndromes Neurotóxicas/diagnóstico , Paclitaxel/uso terapêutico , Polimorfismo Genético , Polimorfismo de Nucleotídeo Único/genética , Espanha
3.
Horm Metab Res ; 41(9): 672-5, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19343621

RESUMO

Hereditary susceptibility to pheochromocytoma (PCC) and paraganglioma (PGL) represents a very complex genetic scenario. It has been reported that the absence of familial antecedents of the disease does not preclude the existence of a mutation affecting any of the five major susceptibility genes. In fact, 11-24% of apparently sporadic cases (without familial or syndromic antecedents) harbor an unexpected germline mutation, but we do not know what is happening in "truly apparently" sporadic patients (i.e., apparently sporadic cases diagnosed with only one tumor). In the present study, we have analyzed 135 apparently sporadic patients developing a single tumor for the five major susceptibility genes: VHL, RET, SDHB, SDHC, and SDHD. Fourteen percent of cases were found to harbor a germline mutation, and only 2.2% of patients were older than 45 years at onset. By taking into account the tumor location and a threshold age at onset of 45 years, we propose a rational scheme for genetic testing. Analyzing VHL and RET genes would be recommended only in young patients developing a single PCC. On the other hand, genetic testing of SDHD should be done in all patients developing an extra-adrenal tumor before the age of 45, and SDHC could be the responsible gene in cases developing a single head and neck tumor, independently of age. Finally, the analysis of SDHB should always be performed because of its association to malignancy and the low penetrance of mutations affecting this gene.


Assuntos
Neoplasias das Glândulas Suprarrenais/genética , Testes Genéticos , Paraganglioma/genética , Feocromocitoma/genética , Adolescente , Neoplasias das Glândulas Suprarrenais/diagnóstico , Adulto , Idoso , Feminino , Mutação em Linhagem Germinativa , Humanos , Masculino , Pessoa de Meia-Idade , Paraganglioma/diagnóstico , Feocromocitoma/diagnóstico , Proteínas Proto-Oncogênicas c-ret/genética , Succinato Desidrogenase/genética , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Adulto Jovem
4.
J Med Genet ; 45(4): 233-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18057081

RESUMO

BACKGROUND: Hereditary susceptibility to familial paraganglioma syndromes is mainly due to mutations in one of six genes, including three of the four genes encoding the subunits of the mitochondrial succinate dehydrogenase complex II. Although prevalence, penetrance and clinical characteristics of patients carrying point mutations affecting the genes encoding succinate dehydrogenase have been well studied, little is known regarding these clinical features in patients with gross deletions. Recently, we found two unrelated Spanish families carrying the previously reported SDHB exon 1 deletion, and suggested that this chromosomal region could be a hotspot deletion area. METHODS: We present the molecular characterisation of this apparently prevalent mutation in three new families, and discuss whether this recurrent mutation is due either to the presence of a founder effect or to a hotspot. RESULTS: The breakpoint analysis showed that all Iberian Peninsular families described harbour the same exon 1 deletion, and that a different breakpoint junction segregates in an affected French pedigree. CONCLUSIONS: After haplotyping the SDHB region, we concluded that the deletion detected in Iberian Peninsular people is probably due to a founder effect. Regarding the clinical characteristics of patients with this alteration, it seems that the presence of gross deletions rather than point mutations is more likely related to abdominal presentations and younger age at onset. Moreover, we found for the first time a patient with neuroblastoma and a germline SDHB deletion, but it seems that this paediatric neoplasia in a pheochromocytoma family is not a key component of this disease.


Assuntos
Proteínas Ferro-Enxofre/genética , Síndromes Neoplásicas Hereditárias/genética , Paraganglioma/genética , Deleção de Sequência , Succinato Desidrogenase/genética , Adolescente , Adulto , Sequência de Bases , Criança , Primers do DNA/genética , Éxons , Feminino , Efeito Fundador , Haplótipos , Humanos , Masculino , Síndromes Neoplásicas Hereditárias/enzimologia , Paraganglioma/enzimologia , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único , Espanha
5.
Endocr Relat Cancer ; 14(3): 645-54, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17914095

RESUMO

Testosterone is essential for the growth and function of the luminal prostate cells, but it is also critical for the development of prostate cancer, which in the majority of the cases derives from luminal cells. Cytochrome P450 3A (CYP3A) enzymes hydroxylate testosterone and dehydroepiandrosterone to less active metabolites, which might be the basis for the association between CYP3A polymorphisms and prostate cancer. However, it is unknown whether the CYP3A enzymes are expressed at relevant levels in the prostate and which polymorphisms could affect this tissue-specific CYP3A activity. Thus, we measured CYP3A4, CYP3A5, CYP3A7, and CYP3A43 mRNA in 14 benign prostatic hyperplasias and ten matched non-tumoral/tumoral prostate samples. We found that CYP3A5 mRNA in non-tumoral prostate tissue was 10% of the average amount of liver samples, whereas the expression of the other CYP3A genes was much lower. Similarly to liver, CYP3A5*3 polymorphism decreased CYP3A5 mRNA content 13-fold. CYP3A5 protein was detected in non-tumoral prostate microsomes by western blot, and immunohistochemistry (IHC) localized CYP3A5 exclusively in the basolateral prostate cells. In contrast to the normal tissue, IHC and RT-PCR showed that tumoral tissue lacked CYP3A5 expression. In conclusion, prostate basolateral cells express high levels of CYP3A5 which dramatically decrease in tumoral tissue. This finding supports an endogenous function of CYP3A5 related to the metabolism of intra-prostatic androgens and cell growth, and that polymorphisms affecting CYP3A5 activity may result in altered prostate cancer risk and aggressiveness.


Assuntos
Carcinoma/genética , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Próstata/metabolismo , Neoplasias da Próstata/genética , Idoso , Idoso de 80 Anos ou mais , Carcinoma/metabolismo , Carcinoma/patologia , Citocromo P-450 CYP3A , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Polimorfismo Genético , Hiperplasia Prostática/genética , Hiperplasia Prostática/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia
6.
Mol Microbiol ; 31(4): 1075-85, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10096076

RESUMO

We have identified and characterized the Igt gene of Bacillus subtilis. The prelipoprotein diacylglycerol transferase enzyme (Lgt) catalyses the first reaction in lipomodification of bacterial lipoproteins. Inactivation of Igt in B. subtilis by a nonsense mutation (prs-11 mutation) or by disruption was shown here to abolish lipomodification of prelipoproteins completely, as well as the cleavage of signal peptide. However, unlike in Gram-negative bacteria, the Igt mutants of B. subtilis were fully viable. In agreement with this observation, studies of two lipoproteins, PrsA and BlaP, indicated that non-lipomodified precursors of these proteins were functional and translocated across the cytoplasmic membrane. However, there was release of both precursors from cells, resulting in a reduced level of the cell-bound form. We have shown that the reduced level of the PrsA lipoprotein, a foldase involved in protein secretion, caused impaired protein secretion, a prominent phenotype of Igt mutants. There was no indication that non-lipomodified PrsA displayed reduced activity.


Assuntos
Bacillus subtilis/metabolismo , Proteínas de Bactérias , Genes Bacterianos , Lipoproteínas/metabolismo , Proteínas de Membrana/metabolismo , Transferases/genética , Bacillus subtilis/genética , Bacillus subtilis/crescimento & desenvolvimento , Mapeamento Cromossômico , Endopeptidases/metabolismo , Lipoproteínas/química , Lipoproteínas/genética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Mutação , Precursores de Proteínas/metabolismo , Subtilisinas/metabolismo , alfa-Amilases/metabolismo
7.
Mol Microbiol ; 31(2): 533-43, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10027970

RESUMO

ecs is a three-cistron operon of Bacillus subtilis, encoding proteins with similarity to the ATPase (EcsA) and hydrophobic components (EcsB) of ABC transporters. The ecsA26 point mutation was shown to cause a strong processing defect of a secreted alpha-amylase precursor (preAmyQ) and of three other exoproteins. Northern analysis of the level of amyQ mRNA showed that ecsA26 also decreases amyQ transcription. This effect too was pleiotropic, as judged by a drastic decrease in the expression from an exoprotease promoter of a reporter protein. A knockout mutation of the ecsB cistron caused a processing defect similar to ecsA26 but, unlike ecsA26, did not affect amyQ transcription. These was also no defect in transcription in the ecsA ecsB double mutant. Thus, an intact ecsB product was required for the downregulation of amyQ by the mutant ecsA. These results suggest a dual regulatory function for Ecs, in which Ecs, possibly as part of a signal transduction mechanism, regulates some component(s) of the protein secretion apparatus as well as secretory protein transcription in a co-ordinated fashion.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Bacillus subtilis/metabolismo , Proteínas de Bactérias/metabolismo , Transdução de Sinais , Transportadores de Cassetes de Ligação de ATP/genética , Bacillus subtilis/genética , Proteínas de Bactérias/genética , Regulação Bacteriana da Expressão Gênica , Óperon , Processamento de Proteína Pós-Traducional , alfa-Amilases
8.
Cytogenet Cell Genet ; 57(1): 9-10, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1713142

RESUMO

We have previously reported the identification and basic characterization of two biallelic TaqI RFLPs, A and B, of the 3' end of the human ACPP locus in an unselected Finnish population (Winqvist et al., 1989). In the present investigation, a similar allelic distribution was observed in patients with prostatic cancer or benign hyperplasia. In addition, it was found that the DNA sequences generating RFLP-B are located further downstream from the RFLP-A sequences.


Assuntos
Fosfatase Ácida/genética , Polimorfismo de Fragmento de Restrição , Próstata/enzimologia , Hiperplasia Prostática/genética , Neoplasias da Próstata/genética , DNA/genética , Sondas de DNA , Desoxirribonucleases de Sítio Específico do Tipo II , Genótipo , Humanos , Masculino , Reação em Cadeia da Polimerase , Hiperplasia Prostática/enzimologia , Neoplasias da Próstata/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...