Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Transl Lung Cancer Res ; 10(1): 314-325, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33569315

RESUMO

BACKGROUND: Rearranged during transfection (RET) rearrangements occur in 1-2% of non-small cell lung cancers (NSCLCs). Alectinib administered at doses of 300 mg and 600 mg twice daily (BID) is approved for. ALK: rearranged NSCLC in Japan and other countries, respectively. Since alectinib has activity against RET, we conducted a phase (P) 1/2 study of alectinib to determine its activity in Japanese patients with. RET: rearranged NSCLC. METHODS: This study was a single-arm, open-label, multi-institutional P1/2 trial. Previously treated patients with RET-rearranged NSCLC, screened by nation-wide network (LC-SCRUM-Japan), were recruited. In P1, alectinib (600 or 450 mg BID) was administered following a 3+3 design and its safety was assessed. During P2, alectinib was administered at the recommended dose (RD) determined in P1. The primary endpoint was the objective response rate (ORR) in RET inhibitor-naïve patients treated with the RD of alectinib. RESULTS: Thirty-four patients were administered alectinib. In cohort 1 (600 mg BID) of P1, we observed 5 dose-limiting toxicities (DLTs), including grade 3 rash and thromboembolic event, in 3 of 6 patients. In cohort 2 (450 mg BID), we observed no DLTs in 3 patients. Pharmacokinetic analysis revealed that AUC0-10 to 600 mg BID was higher than that previously reported in global trials. We determined 450 mg BID as the RD for P2. In 25 RET inhibitor-naïve patients, one achieved an objective response (4%) and 13 achieved disease control at 8 weeks (52%). The median progression-free survival (PFS) was 3.4 months (95% CI, 2.0-5.4), while the median overall survival was 19.0 months (5.4-NE). We observed grade 3 adverse events (AEs) (4%) including pneumonitis in P2. CONCLUSIONS: Alectinib exerts limited activity against RET-rearranged NSCLC. Further investigation to elucidate the mechanisms underlying sensitivity and resistance of RET inhibitors is required to improve outcomes for these patients.

2.
Cancer Sci ; 111(2): 561-570, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31782583

RESUMO

Patients with epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer (NSCLC) harboring BIM deletion polymorphism (BIM deletion) have poor responses to EGFR TKI. Mechanistically, the BIM deletion induces preferential splicing of the non-functional exon 3-containing isoform over the functional exon 4-containing isoform, impairing TKI-induced, BIM-dependent apoptosis. Histone deacetylase inhibitor, vorinostat, resensitizes BIM deletion-containing NSCLC cells to EGFR-TKI. In the present study, we determined the safety of vorinostat-gefitinib combination and evaluated pharmacodynamic biomarkers of vorinostat activity. Patients with EGFR-mutated NSCLC with the BIM deletion, pretreated with EGFR-TKI and chemotherapy, were recruited. Vorinostat (200, 300, 400 mg) was given daily on days 1-7, and gefitinib 250 mg was given daily on days 1-14. Vorinostat doses were escalated based on a conventional 3 + 3 design. Pharmacodynamic markers were measured using PBMC collected at baseline and 4 hours after vorinostat dose on day 2 in cycle 1. No dose-limiting toxicities (DLT) were observed in 12 patients. We determined 400 mg vorinostat as the recommended phase II dose (RP2D). Median progression-free survival was 5.2 months (95% CI: 1.4-15.7). Disease control rate at 6 weeks was 83.3% (10/12). Vorinostat preferentially induced BIM mRNA-containing exon 4 over mRNA-containing exon 3, acetylated histone H3 protein, and proapoptotic BIMEL protein in 11/11, 10/11, and 5/11 patients, respectively. These data indicate that RP2D was 400 mg vorinostat combined with gefitinib in BIM deletion/EGFR mutation double-positive NSCLC. BIM mRNA exon 3/exon 4 ratio in PBMC may be a useful pharmacodynamic marker for treatment.


Assuntos
Proteína 11 Semelhante a Bcl-2/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Gefitinibe/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Vorinostat/administração & dosagem , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/genética , Esquema de Medicação , Receptores ErbB/genética , Feminino , Gefitinibe/farmacocinética , Humanos , Neoplasias Pulmonares/genética , Masculino , Pessoa de Meia-Idade , Mutação , Deleção de Sequência , Análise de Sobrevida , Resultado do Tratamento , Vorinostat/farmacocinética
3.
Nephrol Dial Transplant ; 34(6): 960-969, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30325430

RESUMO

BACKGROUND: This study aimed to investigate the contribution of a proliferation-inducing ligand (APRIL), a member of the tumor necrosis factor (TNF) superfamily implicated in plasma cell survival, to the development of plasma cell-rich lesions in immunoglobulin G4-related disease (IgG4-RD). METHODS: We performed immunohistochemical staining for APRIL with Stalk-1 and Aprily-8 antibodies specifically recognizing APRIL-producing cells and secreted APRIL, respectively, in renal and submandibular lesions of IgG4-RD in comparison with those of Sjögren's syndrome and sialolithiasis. RESULTS: Numerous Stalk-1-positive APRIL-producing cells were detectable in lesions of IgG4-RD. These cells, identified as CD163-positive M2 macrophages, secreted APRIL that distributed close to and even on infiltrating plasma cells. In contrast, APRIL-producing cells and the secreted form of APRIL were rarely detectable in lesions of Sjögren's syndrome or sialolithiasis. Notably, APRIL expression decreased concomitantly with the level of plasma cell infiltration after successful glucocorticoid treatment. CONCLUSIONS: Abundant infiltration into tissue lesions of APRIL-producing M2 macrophages and retention of secreted APRIL in plasma-cell-rich areas support a role for APRIL in the pathogenesis of plasma cell-rich lesions in IgG4-RD.


Assuntos
Doença Relacionada a Imunoglobulina G4/metabolismo , Rim/metabolismo , Macrófagos/metabolismo , Plasmócitos/metabolismo , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Proliferação de Células , Feminino , Humanos , Imunoglobulina G/imunologia , Imuno-Histoquímica , Rim/patologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Cálculos das Glândulas Salivares/imunologia , Síndrome de Sjogren/imunologia , Síndrome de Sjogren/patologia
4.
J Med Invest ; 64(3.4): 317-320, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28955006

RESUMO

BACKGROUND: The rearranged during transfection (RET) fusion gene was discovered as a driver oncogene in 1-2% of non-small cell lung cancers (NSCLCs). Alectinib is an approved anaplastic lymphoma kinase (ALK) inhibitor that may also be effective for RET fusion-positive NSCLC. METHODS/DESIGN: RET fusion-positive NSCLC patients treated with at least one regimen of chemotherapy are being recruited. In step 1, alectinib (600 or 450 mg, twice daily) will be administered following a 3+3 design. The primary endpoint is safety. In step 2, alectinib will be administered at the recommended dose (RD) defined by step 1. The primary endpoint is the response rate of RET inhibitor treatment-naïve patients. CONCLUSION: This is the first study to investigate the safety and preliminary efficacy of alectinib in RET fusion-positive NSCLC patients. If successful, alectinib treatment may lead to substantial and important changes in the management of NSCLC with RET fusion genes. J. Med. Invest. 64: 317-320, August, 2017.


Assuntos
Carbazóis/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Protocolos Clínicos , Neoplasias Pulmonares/tratamento farmacológico , Piperidinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-ret/genética , Quinase do Linfoma Anaplásico , Carcinoma Pulmonar de Células não Pequenas/genética , Humanos , Neoplasias Pulmonares/genética , Receptores Proteína Tirosina Quinases/genética
5.
Clin Kidney J ; 9(1): 69-75, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26798464

RESUMO

BACKGROUND: Uromodulin kidney disease (UKD) is an inherited kidney disease caused by a uromodulin (UMOD) gene mutation. The UMOD gene encodes the Tamm-Horsfall protein (THP), which is the most abundant protein in healthy human urine. Because of its rarity, the incidence of UKD has not been fully elucidated. The purpose of the present study is to clarify the frequency of UKD among patients who underwent renal biopsy. METHODS: Immunostaining for THP was performed for patients <50 years of age with renal insufficiency and hyperuricemia without overt urinalysis abnormality from renal biopsy databases. Serum and urinary THP concentrations were evaluated in available individuals. RESULTS: Fifteen patients were selected for immunostaining from a total of 3787 patients. In three independent patients, abnormal THP accumulation in renal tubular cells was observed. A novel missense A247P UMOD mutation was detected in two of the three patients, including one having a typical family history of familial juvenile hyperuricemic nephropathy. Serum and urinary THP concentrations of all available patients with UMOD A247P mutation were significantly lower than those of controls. CONCLUSIONS: In the present study, UKD was detected in <1 in 1000 subjects who underwent renal biopsies. However, in subjects meeting all of the above criteria, abnormal THP accumulation was detected in 20% (3/15), suggesting that renal biopsy with immunostaining for THP is a good tool for diagnosing UKD. Also, low serum THP concentration detected in the present subjects might be a good diagnostic marker or important in understanding the pathogenesis of UKD.

6.
J Urol ; 187(5): 1876-81, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22425046

RESUMO

PURPOSE: Some nonseminomatous germ cell tumors are resistant to any type of chemotherapy. Control of embryonal carcinoma cells is crucial to manage nonseminomatous germ cell tumors. We established SOX2 targeting therapy in an embryonal carcinoma model. MATERIALS AND METHODS: SOX2 expression was evaluated in a series of testicular germ cell tumor tissue samples. The antitumor effect of SOX2 knockdown was analyzed in vitro and in vivo using an embryonal carcinoma model. RESULTS: In testicular germ cell tumor tissue SOX2 was expressed in the foci of embryonal carcinoma but negative in seminoma and yolk sac tumors. In an embryonal carcinoma model SOX2-siRNA induced apoptotic cell death in vitro and significant growth suppression in vivo. CONCLUSIONS: This study shows the therapeutic potential of SOX2 silencing for embryonal carcinoma. However, further improvements are needed in SOX2-siRNA delivery to the tumor.


Assuntos
Carcinoma Embrionário/metabolismo , Carcinoma Embrionário/terapia , Fatores de Transcrição SOXB1/antagonistas & inibidores , Fatores de Transcrição SOXB1/metabolismo , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/terapia , Animais , Carcinoma Embrionário/patologia , Morte Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Inativação Gênica , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos , RNA Interferente Pequeno/uso terapêutico , Seminoma/metabolismo , Seminoma/patologia , Neoplasias Testiculares/patologia , Transfecção
7.
Mol Carcinog ; 51(9): 711-22, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21809391

RESUMO

Testicular germ cell tumors (TGCTs) have a unique epigenetic profile distinct from that of other types of cancer. To further evaluate epigenetics of TGCTs, this study examines DNA methylation patterns of DNA repetitive elements in TGCTs. Bisulfite genomic sequencing and combined bisulfite restriction analysis (COBRA) were used to analyze the methylation patterns of DNA repetitive elements (LINE1 and Alu repeats) in embryonal carcinoma (EC) derived cell lines, primary TGCT tissues, noncancerous testicular tissues adjacent to TGCTs and cancer cells derived from somatic tissues (testicular malignant lymphoma tissues and renal cell carcinoma cell lines). Through both bisulfite genomic sequencing and COBRA, LINE1 was extensively hypomethylated in both seminomatous and nonseminomatous TGCT tissues as well as EC cell lines. We studied two Alu repeats locating in the 5' end of E-cadherin and XIST by bisulfite genomic sequencing. These two Alu elements were extensively hypomethylated in seminomatous TGCTs, but methylated in nonseminomatous TGCTs, including two EC derived cell lines. This increased unmethylated profile in seminomatous TGCTs was observed also by COBRA for Alu repeats. Although partial demethylation of DNA repetitive elements was observed in cancer cells of somatic tissue origin, the degree of demethylation was more pronounced in TGCTs than in cancer cells of somatic tissue origin. We observed abnormal demethylation of DNA repetitive elements in some of the tissues adjacent to TGCTs. The results indicate that the underlying mechanisms to undergo or maintain demethylation of DNA repetitive sequences differ between TGCTs and cancer cells of somatic tissue origin.


Assuntos
Biomarcadores Tumorais/genética , Metilação de DNA , Perfilação da Expressão Gênica , Linfoma/genética , Neoplasias Embrionárias de Células Germinativas/genética , Sequências Repetitivas de Ácido Nucleico/genética , Neoplasias Testiculares/genética , Adulto , Idoso , Western Blotting , Carcinoma Embrionário/genética , Carcinoma de Células Renais/genética , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA de Neoplasias/genética , Epigênese Genética , Humanos , Neoplasias Renais/genética , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , RNA Interferente Pequeno/genética , Testículo/metabolismo , Células Tumorais Cultivadas , Adulto Jovem
8.
Clin Cancer Res ; 16(10): 2751-9, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20460473

RESUMO

PURPOSE: Testicular germ cell tumors (TGCT) have a unique epigenetic profile distinct from that of other types of cancer. Elucidation of these properties has a potential to identify novel markers for TGCTs. EXPERIMENTAL DESIGN: We conducted comprehensive analysis of DNA methyltransferase (DNMT) gene expression in TGCTs. Based on the expression profiles of DNMT genes in TGCTs, we generated a rabbit polyclonal anti-human DNMT3L antibody. We then studied the role of DNMT3L in TGCTs by the treatment of two embryonal carcinoma (EC) cell lines with a small interfering RNA system. Finally, we evaluated the immunohistochemical detection of DNMT3L in TGCT tissues. We also compared the patterns of DNMT3L immunohistochemistry with those of CD30 and SOX2. RESULTS: Among the DNMT genes, we found that mRNA for DNMT3L was specifically expressed in TGCTs, but neither in normal testicular tissues nor in cancer cells of somatic tissue origin. DNMT3L protein was strongly expressed in two EC cell lines, but not in the cell lines of somatic tissue origin. Transfection of small interfering RNA for DNMT3L significantly reduced DNMT3L expression and resulted in growth suppression and apoptosis in EC cells. Immunohistochemical analysis showed that DNMT3L protein was present only in EC cells, but not in the other types of TGCT components and cancer cells of somatic tissue origin. DNMT3L staining was more prominent and specific than CD30 or SOX2 staining for detecting EC cells. CONCLUSION: DNMT3L is a novel marker and is essential for the growth of human embryonal carcinoma.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma Embrionário/genética , DNA (Citosina-5-)-Metiltransferases/biossíntese , Neoplasias Testiculares/genética , Western Blotting , Carcinoma Embrionário/metabolismo , Separação Celular , DNA (Citosina-5-)-Metiltransferases/genética , Citometria de Fluxo , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Microscopia de Fluorescência , RNA Mensageiro/análise , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Testiculares/metabolismo , Transfecção
9.
Int J Cancer ; 127(2): 474-84, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19921695

RESUMO

We previously demonstrated that Pim-3, a protooncogene with serine/threonine kinase activity, was aberrantly expressed in malignant lesions but not in normal tissues of endoderm-derived organs, including pancreas, liver, colon and stomach. Moreover, aberrantly expressed Pim-3 can prevent tumor cell apoptosis by inactivating a proapoptotic molecule, Bad, and enhancing the expression of an antiapoptotic molecule, Bcl-X(L). These observations prompted us to speculate that a chemical targeting Pim-3 kinase may be a good candidate for a novel type of anticancer drug. Hence, we screened various low-molecule compounds by examining their capacity to inhibit Pim-3 kinase activity in vitro. We observed that some synthetic intermediates of stemonamide can inhibit in vitro activities of Pim-3 kinase and its related kinases, such as Pim-1 and Pim-2. Moreover, these compounds inhibit in vitro cell proliferation of various human pancreatic, hepatocellular and colon cancer cell lines. Furthermore, the compounds can induce apoptosis of human pancreatic cancer cell lines in vitro by reducing the amount of phospho-Ser(112)-Bad, but not total amounts of Bad and Pim-3. Finally, when the compound was administered to nude mice injected with a human pancreatic cancer cell line, it retarded tumor growth by increasing apoptotic cell numbers and decreasing proliferating cell numbers without causing serious adverse effects on blood counts. These observations indicate that the chemicals and its related compounds may be effective for the treatment of tumors of endoderm-derived organs, particularly the pancreas.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Neoplasias do Colo/patologia , Neoplasias Hepáticas/patologia , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas c-pim-1/antagonistas & inibidores , Compostos de Espiro/farmacologia , Animais , Western Blotting , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Estrutura Molecular , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Compostos de Espiro/síntese química , Compostos de Espiro/química , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína de Morte Celular Associada a bcl/metabolismo
10.
Int J Androl ; 30(4): 385-92; discussion 392, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17521367

RESUMO

DNA methylation is the best known and most thoroughly studied epigenetic mechanism. Hypermethylation of CpG islands associated with silencing of tumour suppressor genes or tumour-related genes is a common hallmark of human cancer. The list of tumour-related genes with aberrant hypermethylation in their CpG islands has been increasing. There is also the potential for using DNA methylation profile data as markers for various types of human cancer. In this paper, we review the methylation profile of testicular germ cell tumours (TGCTs). We show that TGCTs have distinctive DNA methylation profiles that differ from those of somatic tissue-derived cancers or somatic tissues. We also discuss the methylation profile of TGCTs in terms of the DNA reprogramming that occurs in primordial germ cells or pre-implantation embryos. Finally, we describe the potential clinical utility of this unique methylation phenotype in TGCTs with regard to developing a novel tumour marker. These data suggest that unmethylated DNA fragments in TGCTs may have diagnostic implications. Further elucidation of epigenetic profiles in TGCTs is expected to provide a new insight into the biology of this disease.


Assuntos
Epigênese Genética , Perfilação da Expressão Gênica , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Testiculares/genética , Fator de Crescimento do Tecido Conjuntivo , Metilação de DNA , Humanos , Proteínas Imediatamente Precoces/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Regiões Promotoras Genéticas
11.
Oncol Rep ; 17(6): 1481-5, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17487408

RESUMO

We previously identified GADD34 (growth arrest and DNA damage protein 34) by screening for genes involved in oncogenic-transformation and/or cellular senescence in Ras-transformed rat F2408 fibroblasts (7EJ-Ras), which exhibit anchorage-independent growth and do not senesce. In the current study, we found that transduction of 7EJ-Ras cells with a retroviral vector expressing GADD34 suppressed their proliferation. Furthermore, we observed that fibroblasts derived from GADD34-knockout mice (GADD34-KO MEFs) did not undergo senescence. Whereas the expression of p21 was decreased in GADD34 KO MEFs, its expression was rescued in these cells by ectopic expression of GADD34 by retroviral transduction. These findings suggest that GADD34 contributes to the regulation of p21 expression, and that it suppresses cellular proliferation through the induction of cellular senescence.


Assuntos
Antígenos de Diferenciação/fisiologia , Proteínas de Ciclo Celular/fisiologia , Senescência Celular/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Neoplasias/patologia , Animais , Antígenos de Diferenciação/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p21/genética , Fibroblastos/metabolismo , Genes ras/genética , Camundongos , Camundongos Knockout , Neoplasias/genética , Proteína Fosfatase 1 , Ratos , Retroviridae/genética , Transdução Genética
12.
Hum Mol Genet ; 15(6): 821-30, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16439445

RESUMO

A common deletion at chromosomal arm 14q32 in human renal cell carcinoma (RCC) prompted us to explore a tumor suppressor gene (TSG) in this region. We report that imprinted DLK1 at 14q32, a regulator of adipocyte differentiation, is a candidate TSG in RCCs. DLK1 expression was lost in 39 out of 50 (78%) primary RCC tissues, whereas expression of DLK1 was maintained in every normal kidney tissue examined. DLK1 was expressed in only one of 15 (7%) RCC-derived cell lines. In order to see the biological significance of DLK1 inactivation in RCCs, we tested the effect of restoration of DLK1 in RCC cell lines, using a recombinant retrovirus containing the gene. Reintroduction of DLK1 into DLK1-null RCC cell lines markedly increased anchorage-independent cell death, anoikis and suppressed tumor growth in nude mice. We then investigated the underlying mechanisms for DLK1 inactivation in RCCs. We found loss of heterozygosity at this region in 12 out of 50 RCC tissues (24%). To explore the role of epigenetic regulation of DLK1 inactivation in RCCs, we conducted methylation analysis of the upstream region and the gene body of DLK1. We could not find a differentially methylated region in either the upstream region or the gene body of DLK1. However, we found that gain of methylation upstream of GTL2, a reciprocal imprinted gene for DLK1, is a critical epigenetic alteration for the inactivation of DLK1 in RCCs. The present data have shown that gain of methylation upstream of the untranslated GTL2 leads to pathological downregulation of DLK1 in RCCs.


Assuntos
Carcinoma de Células Renais/genética , Inativação Gênica , Genes Supressores de Tumor , Impressão Genômica , Peptídeos e Proteínas de Sinalização Intercelular/genética , Neoplasias Renais/genética , Proteínas de Membrana/genética , Mutação , Proteínas/genética , Animais , Proteínas de Ligação ao Cálcio , Carcinoma de Células Renais/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Metilação de DNA , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neoplasias Renais/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , RNA Longo não Codificante
13.
Genes Chromosomes Cancer ; 43(1): 104-12, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15672408

RESUMO

Testicular germ-cell tumors (TGCTs) are pluripotent and display protean histology from the germ-cell stage until embryonal and somatic-cell differentiation. These properties make TGCT a fascinating model for studying germ-cell development and gametogenesis. Methylation patterns specific to cell type (stem cells, germ cells, and somatic tissues) occur throughout the normal development of mice. To shed light on the epigenetic phenotypes among histological subtypes of TGCTs, we investigated the methylation and expression of several cancer testis antigen (CTA) genes (MAGEA1, MAGEA3, and SYCP1) in TGCTs. In the current study, we showed that the 5' ends of MAGEA1 and MAGEA3 on the X chromosome are unmethylated in seminomatous TGCTs, regardless of whether MAGEA1 and MAGEA3 are expressed and are methylated in nonseminomatous TGCTs when expression is absent. These distinctive epigenetic phenotypes of MAGEA1 and MAGEA3 also were observed in pure seminomas and in the seminomatous elements of mixed-type TGCTs. In contrast, the 5' end of SYCP1, on chromosome 1, remained predominantly unmethylated, regardless of expression, in both seminomatous and nonseminomatous TGCTs. This pattern of transcriptional regulation of SYCP1 is similar to that observed for XIST in TGCTs. On the basis of the epigenetic phenotypes of CTA genes, we concluded that, first, consistent unmethylated DNA profiles in seminomatous TGCTs imply that methylation may not be the primary control mechanism of programmed gene expression in seminomatous TGCTs, and, second, that nonseminomatous TGCTs might be midway between seminomatous TGCTs and somatic tissues because gene expression in nonseminomatous TGCTs is regulated by methylation in some genes (MAGEA1 and MAGEA3) but not others (SYCP1 and XIST).


Assuntos
Seminoma/genética , Neoplasias Testiculares/genética , Antígenos de Neoplasias/genética , Metilação de DNA , Primers do DNA , DNA de Neoplasias/genética , Germinoma/genética , Humanos , Masculino , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Oncogene ; 23(36): 6163-9, 2004 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-15195139

RESUMO

The increased risk of several types of cancer in Klinefelter syndrome (47XXY) suggests that the extra X chromosome may be involved in the tumorigenesis associated with this syndrome. Here, we show that cancer cells (PSK-1) derived from a patient with Klinefelter syndrome (47XXY) showing loss of an inactive X chromosome subsequently gained active X chromosomes. We found that this abnormal X chromosome composition in PSK-1 is caused by a loss of an inactive X chromosome followed by multiplication of identical active X chromosomes, not by reactivation of an inactive X chromosome. Furthermore, we extended the characterization of loss-of-inactive X in a series of 22 female-derived cancer cell lines (eight breast cancer cell lines, seven ovarian cancer cell lines, and seven cervical cancer cell lines). The data demonstrate that loss-of-inactive X in the female-derived cancer cells is mainly achieved by loss of an inactive X chromosomes followed by multiplication of an identical active X chromosomes. However, distinctive pathways, including reactivation of an inactive X chromosome, are also involved in the mechanisms for loss-of-inactive X and gain-of-active X in female-derived cancer cells. The biological significance of the loss-of-inactive X and gain-of-active X in the oncogenesis of Klinefelter syndrome and female-derived cancer cells are discussed.


Assuntos
Cromossomos Humanos X , Síndrome de Klinefelter/genética , Neoplasias/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Metilação de DNA , Mecanismo Genético de Compensação de Dose , Feminino , Genes p53 , Humanos , Síndrome de Klinefelter/complicações , Masculino , Neoplasias/metabolismo , Neoplasias Ovarianas/genética , Polimorfismo Genético , RNA Longo não Codificante , RNA não Traduzido/metabolismo , Neoplasias do Colo do Útero/genética
15.
Urology ; 63(4): 773-5, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15072901

RESUMO

Renal hypothermia using ice slush for retroperitoneal laparoscopic partial nephrectomy was performed in 2 patients. A cylindrical device was secured in the enlarged port site. Through the device, ice slush was introduced and placed around the kidney. The nadir renal temperature under renal ischemia in the 2 patients was 18.4 degrees C and 25.8 degrees C.


Assuntos
Hipotermia Induzida/métodos , Gelo , Rim/irrigação sanguínea , Laparoscopia/métodos , Nefrectomia/métodos , Idoso , Temperatura Corporal/fisiologia , Desenho de Equipamento , Humanos , Hipotermia Induzida/instrumentação , Isquemia/etiologia , Neoplasias Renais/cirurgia , Masculino , Pessoa de Meia-Idade , Rim Displásico Multicístico/cirurgia , Nefrectomia/instrumentação , Espaço Retroperitoneal , Instrumentos Cirúrgicos
17.
Lancet ; 363(9402): 40-2, 2004 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-14723995

RESUMO

Testicular germ-cell tumours (TGCTs) are the most common malignant diseases among men aged 20-40 years. We developed a DNA tumour marker for TGCTs based on the unmethylated DNA profile of a neoplasm. The 5' end of the XIST gene is mainly hypomethylated in TGCTs irrespective of XIST expression. Male somatic cells, however, show complete methylation through the CpG sites, including the minimum promoter and XIST-conserved repeats. Identification of a XIST unmethylated fragment in male plasma might be diagnostic for TGCTs.


Assuntos
Germinoma/diagnóstico , Germinoma/genética , Neoplasias Testiculares/diagnóstico , Neoplasias Testiculares/genética , Biomarcadores Tumorais/sangue , Cromossomos Humanos X/genética , Cromossomos Humanos X/metabolismo , Metilação de DNA , DNA de Neoplasias/análise , DNA de Neoplasias/sangue , DNA de Neoplasias/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Germinoma/sangue , Humanos , Masculino , Metilação , RNA Longo não Codificante , RNA não Traduzido/sangue , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Neoplasias Testiculares/sangue
18.
Genes Chromosomes Cancer ; 38(1): 97-101, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12874790

RESUMO

Hypermethylation of tumor-suppressor genes has been implicated in the pathogenesis of human cancers. Although a growing number of genes showing hypermethylation is being reported in human cancer, methylation profiles of tumor-related genes in testicular neoplasms have not been well elucidated. This study was designed to show the methylation profiles of multiple CpG islands in testicular germ cell tumors (TGCTs) in comparison with those in testicular malignant lymphomas. We studied the methylation status of E-cadherin, CDKN2B, CDKN2A, BRCA1, RB1, VHL, RASSF1A, RARB, and GSTP1 by use of TGCT tissues and testicular malignant lymphoma tissues (25 primary TGCT tissues and three primary testicular lymphoma tissues). Methylation was not observed in E-cadherin, CDKN2B, CDKN2A, BRCA1, RB1, VHL, RASSF1A, RARB, and GSTP1 in any of the TGCT tissues. In contrast, all three (100%) of the testicular lymphoma tissues demonstrated hypermethylation of E-cadherin, RASSF1A, and RARB, but not CDKN2B, CDKN2A, BRCA1, RB1, VHL, and GSTP1. These data demonstrate that a distinctive epigenetic phenotype underlies the TGCTs and testicular lymphomas at the CpG sites of E-cadherin, RASSF1A, and RARB; a distinctive epigenetic phenotype was not observed among seminomatous TGCTs and non-seminomatous TGCTs at the CpG sites examined.


Assuntos
Metilação de DNA , Germinoma/genética , Linfoma/genética , Neoplasias Testiculares/genética , Transformação Celular Neoplásica/genética , Ilhas de CpG/genética , Regulação Neoplásica da Expressão Gênica/genética , Germinoma/etiologia , Humanos , Linfoma/etiologia , Masculino , Fenótipo , Seminoma/genética , Neoplasias Testiculares/etiologia
19.
J Urol ; 169(4): 1546-52, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12629412

RESUMO

PURPOSE: An overabundance of X chromosomes in testicular germ cell tumors and the identification of the candidate testicular germ cell tumor susceptibility gene TGCT1 on Xq27 highlight the potential involvement of X chromosomes in testicular germ cell tumor pathogenesis. The current study was designed to shed light on the question whether the multiple X chromosomes in testicular germ cell tumor are active or inactive through a complex mechanism of X chromosomal gain and XIST expression. MATERIALS AND METHODS: We analyzed 4 testicular germ cell tumor derived cell lines and 20 primary testicular germ cell tumor tissues. The number of X chromosomes was determined by fluorescence in situ hybridization using the X chromosome specific probe. The expression patterns of XIST and the 3 X-linked genes androgen receptor (AR), fragile X mental retardation (FMR1 ) and Glypican 3 (GPC3 ) were studied by reverse transcriptase-polymerase chain reaction. Bisulfite genomic sequencing was used to analyze the methylation patterns of the AR, FMR1 and GPC3 genes. The relative expression levels of the 2 X-linked proto-oncogenes ARAF1 and ELK1 were assayed by quantitative reverse transcriptase-polymerase chain reaction. RESULTS: XIST expression was common in seminomatous testicular germ cell tumors (2 of 2 or 100% of seminoma derived cell lines and 10 of 12 or 83% of seminomatous testicular germ cell tumor tissues) but not in nonseminomatous testicular germ cell tumors (0 of 2 or 0% nonseminoma derived cell lines and 2 of 8 or 25% of nonseminomatous testicular germ cell tumor tissues). However, X chromosomal gain was consistently observed in the 2 types of tumors. XIST expression in testicular germ cell tumors and normal testicular parenchyma was not associated with methylation of the AR, FMR1 or GPC3 genes. After determining the expression patterns of AR, FMR1 and GPC3 in testicular germ cell tumor samples we concluded that multiple X chromosomes in testicular germ cell tumors were predominantly hypomethylated and active regardless of XIST expression. The biological significance of excess active X chromosomes in testicular germ cell tumors was suggested by enhanced expression of the 2 X-linked oncogenes ARAF1 and ELK1 in the testicular germ cell tumor derived cell lines. CONCLUSIONS: The current data may suggest the potential oncogenic implications of X chromosomal gain in testicular germ cell tumors.


Assuntos
Cromossomos Humanos X , Neoplasias Embrionárias de Células Germinativas/genética , RNA não Traduzido/genética , Seminoma/genética , Neoplasias Testiculares/genética , Fatores de Transcrição/genética , Metilação de DNA , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Hibridização in Situ Fluorescente , Masculino , Estadiamento de Neoplasias , Neoplasias Embrionárias de Células Germinativas/patologia , RNA Longo não Codificante , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Seminoma/patologia , Aberrações dos Cromossomos Sexuais , Neoplasias Testiculares/patologia , Testículo/patologia , Células Tumorais Cultivadas
20.
Urology ; 61(1): 226-30, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12559313

RESUMO

OBJECTIVES: To analyze the methylation status and expression profiles of multiple tumor suppressor genes in renal cell carcinoma-derived cell lines. Aberrant promoter methylation is commonly found in human cancers. Nonetheless, it is challenging to demonstrate that methylation of a specific gene results in gene inactivation. METHODS: We simultaneously analyzed methylation and expression profiles of five putative tumor suppressor genes (p15, p16, Rb, BRCA1, and E-cadherin) in 14 different cell lines using bisulfite genomic sequencing and reverse transcriptase-polymerase chain reaction. We also used multiplex polymerase chain reaction to identify homozygous deletions at the p15 and p16 loci. RESULTS: Expression of p16, BRCA1, and E-cadherin was maintained in 4 (29%) of 14 cell lines, regardless of the presence of methylation. Aberrant methylation of p16 was observed in 2 (14%), of BRCA1 in 1 (7%), and of E-cadherin in 9 (64%) of 14 cell lines. Concurrent methylation was observed among p16 and BRCA1 (1 [7%] of 14 cell lines) and among p16 and E-cadherin (1 [7%] of 14 cell lines). We detected homozygous deletion of p16 and p15 in 11 (78%) and 6 (43%) cell lines, respectively. CONCLUSIONS: The present data shows the presence of methylation does not always contribute to the loss of expression of tumor suppressor genes. Therefore, we must be cautious in interpreting the results of methylation assays--in particular, detection of methylation by nonquantitative methods. The data also demonstrated that multiple tumor suppressor genes are simultaneously inactivated in renal cell carcinoma-derived cell lines by distinctive mechanisms.


Assuntos
Carcinoma de Células Renais/genética , Genes Supressores de Tumor/fisiologia , Neoplasias Renais/genética , Caderinas/genética , Caderinas/fisiologia , Carcinoma de Células Renais/metabolismo , Metilação de DNA , Deleção de Genes , Regulação Neoplásica da Expressão Gênica/fisiologia , Inativação Gênica/fisiologia , Genes BRCA1/fisiologia , Humanos , Neoplasias Renais/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA