Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Mol Oncol ; 16(3): 683-698, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34482638

RESUMO

Testicular germ cell tumors (TGCTs) are aggressive but sensitive to cisplatin-based chemotherapy. Alternative therapies are needed for tumors refractory to cisplatin with hypomethylating agents providing one possibility. The mechanisms of cisplatin hypersensitivity and resistance in TGCTs remain poorly understood. Recently, it has been shown that TGCTs, even those resistant to cisplatin, are hypersensitive to very low doses of hypomethylating agents including 5-aza deoxy-cytosine (5-aza) and guadecitabine. We undertook a pharmacogenomic approach in order to better understand mechanisms of TGCT hypomethylating agent hypersensitivity by generating a panel of acquired 5-aza-resistant TGCT cells and contrasting these to previously generated acquired isogenic cisplatin-resistant cells from the same parent. Interestingly, there was a reciprocal relationship between cisplatin and 5-aza sensitivity, with cisplatin resistance associated with increased sensitivity to 5-aza and 5-aza resistance associated with increased sensitivity to cisplatin. Unbiased transcriptome analysis revealed 5-aza-resistant cells strongly downregulated polycomb target gene expression, the exact opposite of the finding for cisplatin-resistant cells, which upregulated polycomb target genes. This was associated with a dramatic increase in H3K27me3 and decrease in DNMT3B levels in 5-aza-resistant cells, the exact opposite changes seen in cisplatin-resistant cells. Evidence is presented that reciprocal regulation of polycomb and DNMT3B may be initiated by changes in DNMT3B levels as DNMT3B knockdown alone in parental cells resulted in increased expression of H3K27me3, EZH2, and BMI1, conferred 5-aza resistance and cisplatin sensitization, and mediated genome-wide repression of polycomb target gene expression. Finally, genome-wide analysis revealed that 5-aza-resistant, cisplatin-resistant, and DNMT3B-knockdown cells alter the expression of a common set of polycomb target genes. This study highlights that reciprocal epigenetic changes mediated by DNMT3B and polycomb may be a key driver of the unique cisplatin and 5-aza hypersensitivity of TGCTs and suggests that distinct epigenetic vulnerabilities may exist for pharmacological targeting of TGCTs.


Assuntos
Antineoplásicos , Neoplasias Embrionárias de Células Germinativas , Neoplasias Testiculares , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Metilação de DNA/genética , Resistencia a Medicamentos Antineoplásicos/genética , Epigênese Genética , Humanos , Masculino , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia
2.
Cancer ; 122(12): 1836-43, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27018785

RESUMO

BACKGROUND: Intratumoral heterogeneity presents a major obstacle to the widespread implementation of precision medicine. The authors assessed the origin of intratumoral heterogeneity in nonseminomatous germ cell tumor of the testis (NSGCT) and identified distinct tumor subtypes and a potentially lethal phenotype. METHODS: In this retrospective study, all consecutive patients who had been diagnosed with an NSGCT between January 2000 and December 2010 were evaluated. The histologic makeup of primary tumors and the clinical course of disease were determined for each patient. A Fine and Gray proportional hazards regression analysis was used to determine the prognostic risk factors, and the Gray test was used to detect differences in the cumulative incidence of cancer death. In a separate prospective study, next-generation sequencing was performed on tumor samples from 9 patients to identify any actionable mutations. RESULTS: Six hundred fifteen patients were included in this study. Multivariate analysis revealed that the presence of yolk sac tumor in the primary tumor (P = .0003) was associated with an unfavorable prognosis. NSGCT could be divided into 5 subgroups. Patients in the yolk sac-seminoma subgroup had the poorest clinical outcome (P = .0015). These tumors tended to undergo somatic transformation (P < .0001). Among the 9 NSGCTs that had a yolk sac tumor phenotype, no consistent gene mutation was detected. CONCLUSIONS: The current data suggest that intratumoral heterogeneity is caused in part by differentiation of pluripotent progenitor cells. Integrated or multimodal therapy may be effective at addressing intratumoral heterogeneity and treating distinct subtypes as well as a potentially lethal phenotype of NSGCT. Cancer 2016;122:1836-43. © 2016 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution-NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.


Assuntos
Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/patologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia , Adolescente , Adulto , Idoso , Diferenciação Celular/fisiologia , Criança , Heterogeneidade Genética , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Células-Tronco Neoplásicas/patologia , Fenótipo , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Adulto Jovem
3.
Endocrine ; 45(1): 148-52, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23636804

RESUMO

The presence of the sodium/iodide symporter (NIS) is the prerequisite for the use of the radioiodine in the treatment of thyroid cancer. Thus, stimulators of NIS expression and function are currently investigated in cellular models of various human malignancies, also including extrathyroid cancers. In this study, we analyzed the effects of the histone deacetylase inhibitors (HDACi), suberoylanilide hydroxamic acid (SAHA) and valproic acid (VPA), on NIS expression and function in rat Leydig testicular carcinoma cells (LC540). LC540 cells were exposed to SAHA 3 µM and VPA 3 mM (alone and in combination), and cell viability evaluated by MTT assay and cell counting, NIS mRNA and protein levels by using, respectively, real-time RT-PCR and western blotting. NIS function was evaluated by iodide uptake assay. We found that both HDACi were able to stimulate the transcription of NIS gene, but not its protein expression, while the association of SAHA and VPA increased both NIS transcript and protein levels, resulting in significant sixfold enhancement of radioiodine uptake capacity of LC540 cells. These data demonstrate the presence of an epigenetic control of NIS expression in Leydig tumor cells, suggesting the possibility to use the combination of these two HDACi for a radioiodine-based treatment of these malignancies.


Assuntos
Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Tumor de Células de Leydig/patologia , Simportadores/genética , Neoplasias Testiculares/patologia , Ácido Valproico/farmacologia , Animais , Linhagem Celular Tumoral , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Expressão Gênica/efeitos dos fármacos , Tumor de Células de Leydig/tratamento farmacológico , Tumor de Células de Leydig/genética , Masculino , Ratos , Simportadores/fisiologia , Neoplasias Testiculares/tratamento farmacológico , Neoplasias Testiculares/genética , Vorinostat
4.
J Steroid Biochem Mol Biol ; 136: 238-46, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23098692

RESUMO

The active form of vitamin D, 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) has anti-proliferative, pro-apoptotic, and pro-differentiating effects in somatic cancer cells in vitro and in vivo. 1,25(OH)2D3 also augments the anti-tumor effects of several chemotherapeutic agents, including cisplatin, which may have clinical relevance. Given the pro-differentiation effect of vitamin D recently demonstrated in testicular germ cell tumors (TGCTs), we hypothesized that 1,25(OH)2D3 could be a beneficial adjunctive to existing chemotherapy regime used to treat these tumors. In this study, cell survival effects of 1,25(OH)2D3, another pro-differentiation compound, retinoic acid and cisplatin were investigated in TGCT-derived cell lines in vitro. 1,25(OH)2D3 augmented the effect of cisplatin in an embryonal carcinoma-derived cell line (NTera2), possibly through downregulation of pluripotency genes and simultaneous upregulation of the cell cycle regulators p21, p27, p53, p73 and FOXO1, while no significant effects were found in TCam-2 and 2102Ep cell lines (derived from seminoma and embryonal carcinoma, respectively). Anti-tumor effects of cholecalciferol, 1,25(OH)2D3, and cisplatin were subsequently tested in vivo, in a NTera2 xenograft tumor model in nude mice. In xenograft tumors, co-treatment with 1,25(OH)2D3 and cisplatin resulted in downregulation of OCT4 and simultaneous upregulation of p21 and p73, but did not reduce tumor growth significantly more than cisplatin alone. Also, cholecalciferol supplemented diet (1100IU daily) after tumor formation did not increase cisplatin sensitivity in vivo. In conclusion, addition of 1,25(OH)2D3 augmented the antitumor effect of cisplatin monotherapy in vitro, but not in this in vivo testicular germ cell cancer model. Future studies are needed to investigate potential beneficial effects of vitamin D with lower cisplatin doses, and to determine whether 1,25(OH)2D3 may increase cisplatin sensitivity in chemotherapy-resistant TGCTs. This article is part of a Special Issue entitled 'Vitamin D Workshop'.


Assuntos
Antineoplásicos/administração & dosagem , Calcitriol/administração & dosagem , Cisplatino/administração & dosagem , Resistencia a Medicamentos Antineoplásicos , Neoplasias Embrionárias de Células Germinativas/tratamento farmacológico , Neoplasias Testiculares/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Linhagem Celular Tumoral , Suplementos Nutricionais , Sinergismo Farmacológico , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/patologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia
6.
Int J Mol Med ; 13(5): 685-90, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15067371

RESUMO

Therapeutic procedures in patients with testicular germ cell tumors (GCT) are determined by the histopathology of the primary tumor and the tumor extension. The aim of our study was to determine whether conventional staging could be supplemented by combining enrichment of disseminated testicular GCT cells from peripheral blood with subsequent detection of germ-cell-specific gene products. Blood samples from 46 patients with GCT of different clinical stages (CS) were examined by RT-PCR before therapy and >/=8 weeks thereafter for alpha-fetoprotein, beta-human chorionic gonadotropin and germ-cell-specific alkaline phosphatase mRNA. In addition, we performed titration experiments to evaluate whether the sensitivity can be improved by previous immunomagnetic tumor cell enrichment with anti-epithelial HEA-125 microbeads. No positive results were found in controls (n=15; specificity 100%). The overall ratio of positive PCR results in the group of patients with GCTs was 28.26%. The ratio was 35.7% for CS >IIb (n=5/14 patients), 20.0% for CS IIa-b (n=4/20) and 33.3% for CS I (n=4/12). FACS analysis in titration experiments with GCT cell lines showed that previous immunomagnetic tumor cell enrichment achieved a significant increase ranging up to 185.6 times the initial ratio and thus improved the measuring conditions for detection of tumor-specific transcripts. The sole qualitative RT-PCR of tumor-specific gene products in peripheral blood is not sensitive enough to improve staging in GCT patients. Immunomagnetic enrichment of GCT cells in peripheral blood seems a promising approach for increasing the sensitivity of RT-PCR.


Assuntos
Germinoma/sangue , Germinoma/genética , Separação Imunomagnética/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Neoplasias Testiculares/sangue , Neoplasias Testiculares/genética , Proteínas de Ligação ao Cálcio/sangue , Proteínas de Ligação ao Cálcio/genética , Gonadotropina Coriônica Humana Subunidade beta/sangue , Gonadotropina Coriônica Humana Subunidade beta/genética , Proteínas de Ligação a DNA/sangue , Citometria de Fluxo , Germinoma/patologia , Proteínas Ativadoras de Guanilato Ciclase , Humanos , Masculino , Estadiamento de Neoplasias , RNA Mensageiro/sangue , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Neoplasias Testiculares/patologia , alfa-Fetoproteínas
7.
Gene ; 323: 189-99, 2003 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-14659893

RESUMO

The human TPTE (Transmembrane Phosphatase with TEnsin homology) gene family encodes a PTEN-related tyrosine phosphatase with four potential transmembrane domains. Chromosomal mapping revealed multiple copies of the TPTE gene on chromosomes 13, 15, 21, 22 and Y. Human chromosomes 13 and 21 copies encode two functional proteins, TPIP (TPTE and PTEN homologous Inositol lipid Phosphatase) and TPTE, respectively, whereas only one copy of the gene exists in the mouse genome. In the present study, we show that TPTE and TPIP proteins are expressed in secondary spermatocytes and/or prespermatids. In addition, we report the existence of several novel alternatively spliced isoforms of these two proteins with variable number of transmembrane domains. The latter has no influence on the subcellular localization of these different peptides as shown by co-immunofluorescence experiments. Finally, we identify another expressed TPTE copy, mapping to human chromosome 22, whose transcription appears to be under the control of the LTR of human endogenous retrovirus RTVL-H3.


Assuntos
Processamento Alternativo , Proteínas de Membrana/genética , Proteínas Tirosina Fosfatases/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Linhagem Celular , Chlorocebus aethiops , Cromossomos Humanos Par 22/genética , DNA Complementar/química , DNA Complementar/genética , Feminino , Regulação Enzimológica da Expressão Gênica , Proteínas de Fluorescência Verde , Células HeLa , Humanos , Imuno-Histoquímica , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutação , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Pseudogenes/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Análise de Sequência de DNA , Espermátides/enzimologia , Espermatócitos/enzimologia , Neoplasias Testiculares/enzimologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/patologia , Testículo/enzimologia
8.
Oncogene ; 16(1): 95-103, 1998 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-9467948

RESUMO

We describe the cloning and initial characterization of a novel cDNA from human embryonal carcinoma (EC) cells. This cDNA, which we named human growth differentiation factor 3 (hGDF3), encodes the homologue of mouse GDF3, a TGFbeta superfamily member belonging to the Growth/Differentiation Factors. We have analysed the expression of hGDF3 in human embryonal carcinoma cell lines and in primary testicular germ cell tumours of adolescents and adults (TGCTs). Expression of hGDF3 in human EC cell lines is stem cell-specific, is down-regulated upon RA-mediated differentiation and is increased upon culture of the cells in the presence of activin A. In TGCTs, hGDF3 expression is low in seminomas, while expression in non-seminomas is readily detectable and appears to be associated with the EC and yolk sac components in the tumours. We have also mapped the hGDF3 locus to the short arm of human chromosome 12, a region consistently overrepresented in human testicular germ cell tumours. Thus, hGDF3 represents an embryonal carcinoma stem cell-associated marker both in vitro and in vivo.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Substâncias de Crescimento/genética , Peptídeos e Proteínas de Sinalização Intercelular , Teratoma/genética , Neoplasias Testiculares/genética , Ativinas , Sequência de Aminoácidos , Sequência de Bases , Fragmentação do DNA , DNA Complementar , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Fator 3 de Diferenciação de Crescimento , Humanos , Inibinas/farmacologia , Masculino , Dados de Sequência Molecular , RNA Mensageiro/genética , Homologia de Sequência de Aminoácidos , Teratoma/patologia , Neoplasias Testiculares/patologia , Tretinoína/farmacologia , Células Tumorais Cultivadas
9.
Pharmacogenetics ; 7(1): 21-5, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9110358

RESUMO

The incidence rate of testicular cancer has been steadily increasing during the last 50 years, and only cryptorchidism, i.e. undescended testes, has been identified as an important risk factor. An interplay between changing environmental factors and genetic susceptibility e.g. in foreign compound metabolizing enzymes, may have important influences on the risk. The aim of this study was to investigate if glutathione S-transferase mu (GST mu) deficiency, which in previous studies has been associated with malignant melanoma and cancers of the lung and bladder, is a risk factor of testicular cancer. Three hundred and seventy-eight men participated (80 seminomas, 104 non-seminomas and 194 controls) in a population-based case-control study. The phenotype of GST mu was determined in 366 men by ELISA, the genotype was determined in 324 men by polymerase chain reaction. The concordance between geno- and phenotype was 94.4%. The odds ratio of having the GST mu negative phenotype and testicular cancer was 1.08, (0.72-1.64; 95% confidence interval (CI)), and the odds ratio of having the GSTM1 null genotype and testicular cancer was 1.10; CI95% (0.71-1.70). This study provides no evidence of an association between phenotypically determined GST mu deficiency or GSTM1 null genotype and testicular cancer. The narrow confidence intervals rule out GST mu as a major single risk factor for testicular cancer.


Assuntos
Glutationa Transferase/genética , Isoenzimas/genética , Neoplasias Testiculares/genética , Adulto , Estudos de Casos e Controles , Café , Criptorquidismo/epidemiologia , Dinamarca , Exercício Físico , Genótipo , Humanos , Incidência , Masculino , Fenótipo , Seminoma/enzimologia , Seminoma/genética , Fumar , Neoplasias Testiculares/enzimologia , Neoplasias Testiculares/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA