Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
1.
Oncogene ; 34(11): 1463-74, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-24704825

RESUMO

LNK (SH2B3) is an adaptor protein studied extensively in normal and malignant hematopoietic cells. In these cells, it downregulates activated tyrosine kinases at the cell surface resulting in an antiproliferative effect. To date, no studies have examined activities of LNK in solid tumors. In this study, we found by in silico analysis and staining tissue arrays that the levels of LNK expression were elevated in high-grade ovarian cancer. To test the functional importance of this observation, LNK was either overexpressed or silenced in several ovarian cancer cell lines. Remarkably, overexpression of LNK rendered the cells resistant to death induced by either serum starvation or nutrient deprivation, and generated larger tumors using a murine xenograft model. In contrast, silencing of LNK decreased ovarian cancer cell growth in vitro and in vivo. Western blot studies indicated that overexpression of LNK upregulated and extended the transduction of the mitogenic signal, whereas silencing of LNK produced the opposite effects. Furthermore, forced expression of LNK reduced cell size, inhibited cell migration and markedly enhanced cell adhesion. Liquid chromatography-mass spectroscopy identified 14-3-3 as one of the LNK-binding partners. Our results suggest that in contrast to the findings in hematologic malignancies, the adaptor protein LNK acts as a positive signal transduction modulator in ovarian cancers.


Assuntos
Proteínas 14-3-3/metabolismo , Proliferação de Células/fisiologia , Neoplasias Ovarianas/patologia , Proteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Tamanho Celular , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transplante de Neoplasias , Ligação Proteica , Proteínas/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Transplante Heterólogo
2.
Br J Cancer ; 109(7): 1795-804, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24002598

RESUMO

BACKGROUND: The PIAS4 protein belongs to the family of protein inhibitors of activated STAT, but has since been implicated in various biological activities including the post-translational modification known as sumoylation. In this study, we explored the roles of PIAS4 in pancreatic tumourigenesis. METHODS: The expression levels of PIAS4 in pancreatic cancer cells were examined. Cell proliferation and invasion was studied after overexpression and gene silencing of PIAS4. The effect of PIAS4 on hypoxia signalling was investigated. RESULTS: The protein was overexpressed in pancreatic cancer cells compared with the normal pancreas. Gene silencing by PIAS4 small interfering RNA (siRNA) suppressed pancreatic cancer cell growth and overexpression of PIAS4 induced expression of genes related to cell growth. The overexpression of PIAS4 is essential for the regulation of the hypoxia signalling pathway. PIAS4 interacts with the tumour suppressor von Hippel-Lindau (VHL) and leads to VHL sumoylation, oligomerization, and impaired function. Pancreatic cancer cells (Panc0327, MiaPaCa2) treated with PIAS4 siRNA suppressed expression of the hypoxia-inducible factor hypoxia-inducible factor 1 alpha and its target genes JMJD1A, VEGF, and STAT3. CONCLUSION: Our study elucidates the role of PIAS4 in the regulation of pancreatic cancer cell growth, where the suppression of its activity represents a novel therapeutic target for pancreatic cancers.


Assuntos
Hipóxia Celular , Neoplasias Pancreáticas/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Adenocarcinoma/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Histona Desmetilases com o Domínio Jumonji/biossíntese , Proteínas de Ligação a Poli-ADP-Ribose , Proteínas Inibidoras de STAT Ativados/genética , Interferência de RNA , RNA Interferente Pequeno , Fator de Transcrição STAT3/biossíntese , Transdução de Sinais , Sumoilação , Fator A de Crescimento do Endotélio Vascular/biossíntese
3.
Oncogenesis ; 2: e47, 2013 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-23689287

RESUMO

Approximately 90% of well-differentiated/de-differentiated liposarcomas (WDLPS/DDLPS), the most common LPS subtype, have chromosomal amplification at 12q13-q22. Many protein-coding genes in the region, such as MDM2 and , have been studied as potential therapeutic targets for LPS treatment, with minimal success. In the amplified region near the MDM2 gene, our single nucleotide polymorphism (SNP) array analysis of 75 LPS samples identified frequent amplification of miR-26a-2. Besides being in the amplicon, miR-26a-2 was overexpressed significantly in WDLPS/DDLPS (P<0.001), as well as in myxoid/round cell LPS (MRC) (P<0.05). Furthermore, Kaplan-Meier survival analysis showed that overexpression of miR-26a-2 significantly correlated with poor patient survival in both types of LPS (P<0.05 for WDLPS/DDLPS; P<0.001 for MRC). Based on these findings, we hypothesized that miR-26a-2 has an important role in LPS tumorigenesis, regardless of LPS subtypes. Overexpression of miR-26a-2 in three LPS cell lines (SW872, LPS141 and LP6) enhanced the growth and survival of these cells, including faster cell proliferation and migration, enhanced clonogenicity, suppressed adipocyte differentiation and/or resistance to apoptosis. Inhibition of miR-26a-2 in LPS cells using anti-miR-26a-2 resulted in the opposite responses. To explain further the effect of miR-26a-2 overexpression in LPS cells, we performed in silico analysis and identified 93 candidate targets of miR-26a-2. Among these genes, RCBTB1 (regulator of chromosome condensation and BTB domain-containing protein 1) is located at 13q12.3-q14.3, a region of recurrent loss of heterozygosity (LOH) in LPS. Indeed, either overexpression or inhibition of RCBTB1 made LPS cells more susceptible or resistant to apoptosis, respectively. In conclusion, our study for the first time reveals the contribution of miR-26a-2 to LPS tumorigenesis, partly through inhibiting RCBTB1, suggesting that miR-26a-2 is a novel therapeutic target for human LPS.

4.
Oncogene ; 32(9): 1155-63, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-22580610

RESUMO

Chromosome 1p36.23 is frequently deleted in glioblastoma multiforme (GBM). miR-34a localizes in this region. Our experiments found that miR-34a was often deleted and epidermal growth factor receptor (EGFR) was frequently amplified in genomic DNA of 55 GBMs using single-nucleotide polymorphism DNA microarray. Notably, we found that the mean survival time was significantly shortened for patients whose GBMs had both EGFR amplification and miR-34a deletion. Expression of miR-34a was significantly lower in GBM samples compared with normal brain tissue. Forced expression of miR-34a in GBM cells decreased their ability to migrate and profoundly decreased their levels of cyclin-A1, -B1, -D1, and -D3, as well as cyclin-dependent kinase and increased expression of cyclin kinase inhibitor proteins (p21, p27). Also, human GBM cells (U251) stable overexpressing mir-34a formed smaller tumors when growing as xenografts in immunodeficient mice compared with wild-type U251 GBM cells. Furthermore, the protein expression of EGFR decreased in the cells with forced overexpression of miR-34a. Additional studies showed that mir-34a targeted Yin Yang-1 (YY1) and YY1 is a transcription factor that can stimulate the expression of EGFR. Thus, our data suggest that miR-34a acts as a tumor suppressor by inhibiting growth of GBM cells in vitro and in vivo associated with moderating the expression of cell-cycle proteins and EGFR. Moreover, we discovered for the first time that both deletion of miR-34a and amplification of EGFR were associated with significantly decreased overall survival of GBM patients.


Assuntos
Neoplasias Encefálicas/genética , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , MicroRNAs/fisiologia , Animais , Neoplasias Encefálicas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Amplificação de Genes , Deleção de Genes , Genes Supressores de Tumor , Glioblastoma/mortalidade , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Nus , Transplante Heterólogo , Fator de Transcrição YY1/metabolismo
5.
Oncogene ; 25(13): 1852-61, 2006 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-16288219

RESUMO

DLK1 (delta-like) is a transmembrane and secreted protein in the epidermal growth factor-like homeotic family. Although expressed widely during embryonic development, only a few tissues retain the expression in adults. Neuroendocrine tumors often highly express this protein; therefore, we hypothesized that brain tumors might also express it. This study found that the expression of DLK1 in gliomas was higher than that in normal brain (P < 0.05). After stable transfection of a DLK1 cDNA expression vector into GBM cell lines, their proliferation was increased. Furthermore, they lost contact inhibition, had enhanced anchorage-independent growth in soft agar, and had significantly greater capacity to migrate. Western blot studies showed that expression of cyclin D1, CDK2, and E2F4 were increased, and Rb levels were decreased in these cells. DLK1 was found on the cell surface and secreted in the medium from the transfected GBM cells. DLK1-enriched condition medium stimulated the growth of glioblastoma multiforme cell lines and explants. DLK1 antibody blocked cell growth stimulated by DLK1. In summary, these results suggest that DLK1 may play a role in the formation or progression of gliomas.


Assuntos
Neoplasias Encefálicas/genética , Transformação Celular Neoplásica/genética , Glioma/genética , Proteínas de Membrana/biossíntese , Proteínas Repressoras/biossíntese , Western Blotting , Neoplasias Encefálicas/fisiopatologia , Proteínas de Ligação ao Cálcio , Movimento Celular , Proliferação de Células , DNA Complementar/biossíntese , Progressão da Doença , Perfilação da Expressão Gênica , Glioma/fisiopatologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Células Tumorais Cultivadas
6.
Leuk Lymphoma ; 42(3): 517-20, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11699418

RESUMO

The CHK2 gene encodes a protein kinase that is important for the regulation of cell cycle arrest after DNA damage. CHK2 acts downstream of ataxia teleangiecstasia mutated (ATM), modulates the function of p53 and may help mediate cell cycle arrest at G2/M by phosphorylation of Cdc25C. Recently, the human homolog of the checkpoint kinase Cds1 (CHK2) has been suggested to be a tumor suppressor gene. Heterozygous germline mutations have been reported in Li-Fraumeni syndrome (LFS), a highly penetrant familial cancer phenotype, and in sporadic colon cancer. LFS is associated with the development of lymphoid malignancies, especially childhood ALL. Therefore, we analyzed the DNA from 143 lymphoid malignancies to determine whether they had mutations of the CHK2 gene. The 14 exons of CHK2 were studied by polymerase chain reaction-single strand conformational polymorphism (PCR-SSCP) and sequencing of aberrantly migrating bands. One missense mutation changing serine to phenylalanine (codon 428) in an evolutionarily highly conserved domain was found in a non-Hodgkin's aggressive lymphoma. Another point mutation in the non-coding region was identified in one of adult T-cell leukemias (ATL) samples. This result suggests that mutation of the CHK2 gene may rarely be involved in the development of selected lymphomas.


Assuntos
Replicação do DNA/genética , Linfoma não Hodgkin/genética , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases , Adulto , Substituição de Aminoácidos , Ciclo Celular/genética , Quinase do Ponto de Checagem 2 , Criança , Códon , Humanos , Mutação , Polimorfismo Genético , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Homologia de Sequência de Aminoácidos
7.
J Urol ; 166(1): 54-8, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11435822

RESUMO

PURPOSE: We analyzed the effects of the change in TNM classification from the 1987 to the 1997 version and suggest a modified tumor size cutoff point between T stages 1 and 2 for renal cell carcinoma. MATERIALS AND METHODS: We evaluated a database containing the records of 661 patients who underwent nephrectomy between 1989 and 1999. The effect of the change in TNM classification on the distribution of patients between stages, the rates of M+ and N+ disease, and the local and distant recurrence rates were outlined for 280 patients with T stages 1 and 2 disease. The Cox model was used to identify the optimal cutoff point between T1 and T2 disease, and the resulting effect of adopting this cutoff was outlined. RESULTS: A total of 174 and 128 cases were down staged from 1987 version stage T2 to 1997 version stage T1 and from 1987 TNM stage II to 1997 TNM stage I, respectively. Survival was not significantly different in patients with 1997 TNM stages I and II disease due to a lack of survival difference during the first 2 years of followup. Stage shift also caused an increase in average tumor size, the proportion of patients with high grade cancer, and M+ and N+ disease at diagnosis in 1997 stages T1 and T2 as well as an increase in the proportion of 1997 stage T2N0M0 cases at diagnosis with systemic failure. Analysis of 11 potential cutoff points between 1 and 10 cm. revealed that 4.5 cm. was most predictive of patients survival (hazards ratio 4.99, p = 0.0001). Using this cutoff resulted in improved discriminatory power of the TNM classification and a moderating effect on the distribution of patients, average tumor size, high grade disease, M+ and N+ disease at diagnosis, and systemic failure between T(14.5) and T(24.5) compared with 1997 T1 and T2. CONCLUSIONS: Our data imply that the current cutoff point of 7 cm. between stages T1 and T2 tumors is too high. Lowering the cutoff to 4.5 cm. resulted in better discriminatory power of the TNM classification in our dataset. This observation should be further validated by external data.


Assuntos
Carcinoma de Células Renais/mortalidade , Carcinoma de Células Renais/patologia , Neoplasias Renais/mortalidade , Neoplasias Renais/patologia , Estadiamento de Neoplasias/classificação , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Renais/cirurgia , Estudos de Coortes , Intervalo Livre de Doença , Feminino , Humanos , Neoplasias Renais/cirurgia , Masculino , Pessoa de Meia-Idade , Nefrectomia/métodos , Prognóstico , Sensibilidade e Especificidade , Análise de Sobrevida
8.
Blood ; 98(3): 787-94, 2001 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-11468180

RESUMO

An imbalance between cellular apoptosis and survival may be critical for the pathogenesis of lymphoma. Therefore, the gene expression pattern in lymph node preparations from patients with mantle cell lymphoma (MCL) was compared to the pattern in nonmalignant hyperplastic lymph nodes (HLs). Oligonucleotide microarray analysis was performed comparing 5 MCLs to 4 HLs using high-density microarrays. The expression data were analyzed using Genespring software. For confirmation, the expression of selected genes was analyzed by real-time polymerase chain reaction using the RNA extracted from 16 MCL and 12 HL samples. The focus was on 42 genes that were at least 3-fold down-regulated in MCL; in addition to the B-cell leukemia 2 (BCL2) system other apoptotic pathways were altered in MCL. The FAS-associated via death domain (FADD) gene that acts downstream of the FAS cascade as a key gene to induce apoptosis was more than 10-fold down-regulated in MCL. Furthermore, the death-associated protein 6 (DAXX) gene, the caspase 2 (CASP2) gene, and the RIPK1 domain containing adapter with death domain (RAIDD) gene, which are key genes in other proapoptotic pathways, were also decreased in the MCL samples. The suggestion is made that in addition to the known overexpression of cyclin D1, which drives entry into the cell cycle, disturbances of pathways associated with apoptosis contribute to the development of MCL. (Blood. 2001;98:787-794)


Assuntos
Apoptose/genética , Linfoma de Célula do Manto/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Transdução de Sinais/genética , Apoptose/fisiologia , Ciclina D1/genética , Perfilação da Expressão Gênica , Genes bcl-2 , Genes cdc , Humanos , Linfonodos/patologia , Linfoma de Célula do Manto/genética
9.
Oncogene ; 20(25): 3301-5, 2001 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-11423979

RESUMO

Aneuploidy is a characteristic of the majority of human cancers, and recent studies suggest that defects of mitotic checkpoints play a role in carcinogenesis. MAD1L1 is a checkpoint gene, and its dysfunction is associated with chromosomal instability. Rare mutations of this gene have been reported in colon and lung cancers. We examined a total of 44 cell lines (hematopoietic, prostate, osteosarcoma, breast, glioblastoma and lung) and 133 fresh cancer cells (hematopoietic, prostate, breast and glioblastoma) for alterations of MAD1L1 by RT-PCR-SSCP and nucleotide sequencing. Eight mutations consisting of missense, nonsense and frameshift mutations were found, together with a number of nucleotide polymorphisms. All the alterations in cell lines were heterozygous. Frequency of mutations was relatively high in prostate cancer (2/7 cell lines and 2/33 tumor specimens). We placed a mutant truncated MAD1L1, found in a lymphoma sample, into HOS, Ht161 and SJSA cell lines and found that it was less inhibitory than wild type MAD1L1 at decreasing cell proliferation. Co-expression experiments showed that the mutant form had a dominant-negative effect. Furthermore, this mutant impaired the mitotic checkpoint as shown by decreased mitotic indices in HOS cells expressing mutant MAD1L1 after culture with the microtubule-disrupting agent, nocodazole. Our results suggest a pathogenic role of MAD1L1 mutations in various types of human cancer.


Assuntos
Transformação Celular Neoplásica/genética , Mitose/genética , Mutação , Proteínas de Neoplasias/genética , Neoplasias da Mama/genética , Códon sem Sentido , Feminino , Mutação da Fase de Leitura , Neoplasias Hematológicas/genética , Humanos , Neoplasias Pulmonares/genética , Masculino , Mutação de Sentido Incorreto , Osteossarcoma/genética , Neoplasias da Próstata/genética
10.
Lab Invest ; 81(4): 555-64, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11304575

RESUMO

Activation of the TCL1 oncogene has been implicated in T cell leukemias/lymphomas and recently was associated with AIDS diffuse large B cell lymphomas (AIDS-DLBCL). Also, in nonmalignant lymphoid tissues, antibody staining has shown that mantle zone B cells expressed abundant Tcl1 protein, whereas germinal center (GC; centrocytes and centroblasts) B cells showed markedly reduced expression. Here, we analyze isolated B cell subsets from hyperplastic tonsil to determine a more precise pattern of Tcl1 expression with development. We also examine multiple B cell lines and B lymphoma patient samples to determine whether different tumor classes retain or alter the developmental pattern of expression. We show that TCL1 expression is not affected by Epstein-Barr virus (EBV) infection and is high in naïve B cells, reduced in GC B cells, and absent in memory B cells and plasma cells. Human herpesvirus-8 infected primary effusion lymphomas (PEL) and multiple myelomas are uniformly TCL1 negative, whereas all other transformed B cell lines tested express moderate to abundant TCL1. This observation supports the hypothesis that PEL, like myeloma, usually arise from post-GC stages of B cell development. Tcl1 protein is also detected in most naïve/GC-derived B lymphoma patient samples (23 of 27 [85%] positive), whereas most post-GC-derived B lymphomas lack expression (10 of 41 [24%] positive). These data indicate that the pattern of Tcl1 expression is distinct between naïve/GC and post-GC-derived B lymphomas (P < 0.001) and that the developmental pattern of expression is largely retained. However, post-GC-derived AIDS-DLBCL express TCL1 at a frequency equivalent to naïve/GC-derived B lymphomas in immune-competent individuals (7 of 9 [78%] positive), suggesting that TCL1 down-regulation is adversely affected by severe immune system dysfunction. These findings demonstrate that TCL1 expression in B cell lymphoma usually reflects the stage of B cell development from which they derive, except in AIDS-related lymphomas.


Assuntos
Subpopulações de Linfócitos B/metabolismo , Linfoma de Células B/genética , Proteínas Proto-Oncogênicas/genética , Linhagem Celular Transformada , Transformação Celular Viral , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Herpesvirus Humano 4/patogenicidade , Humanos , Hiperplasia/genética , Hiperplasia/metabolismo , Linfoma Relacionado a AIDS/genética , Linfoma Relacionado a AIDS/metabolismo , Linfoma de Células B/classificação , Linfoma de Células B/metabolismo , Linfoma Difuso de Grandes Células B/metabolismo , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Tonsila Palatina/imunologia , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-6 , RNA Mensageiro/biossíntese , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas
11.
J Biol Chem ; 276(17): 14187-94, 2001 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-11297518

RESUMO

To identify genes involved in breast cancer, polymerase chain reaction-selected cDNA subtraction was utilized to construct a breast cancer-subtracted library. Differential screening of the library isolated the growth factor-inducible immediate-early gene Cyr61, a secreted, cysteine-rich, heparin binding protein that promotes endothelial cell adhesion, migration, and neovascularization. Northern analysis revealed that Cyr61 was expressed highly in the invasive breast cancer cell lines MDA-MB-231, T47D, and MDA-MB-157; very low levels were found in the less tumorigenic MCF-7 and BT-20 breast cancer cells and barely detectable amounts were expressed in the normal breast cells, MCF-12A. Univariate analysis showed a significant or borderline significant association between Cyr61 expression and stage, tumor size, lymph node positivity, age, and estrogen receptor levels. Interestingly, expression of Cyr61 mRNA increased 8- to 12-fold in MCF-12A and 3- to 5-fold in MCF-7 cells after 24- and 48-h exposure to estrogen, respectively. Induction of Cyr61 mRNA was blocked by tamoxifen and ICI182,780, inhibitors of the estrogen receptor. Stable expression of Cyr61 cDNA under the regulation of a constitutive promoter in MCF-7 cells enhanced anchorage-independent cell growth in soft agar and significantly increased tumorigenicity and vascularization of these tumors in nude mice. Moreover, overexpression of Cyr61 in MCF-12A normal breast cells induced their tumor formation and vascularization in nude mice. In summary, these results suggest that Cyr61 may play a role in the progression of breast cancer and may be involved in estrogen-mediated tumor development.


Assuntos
Neoplasias da Mama/metabolismo , Estradiol/análogos & derivados , Estrogênios/metabolismo , Substâncias de Crescimento/biossíntese , Proteínas Imediatamente Precoces/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Animais , Antineoplásicos Hormonais/farmacologia , Northern Blotting , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/tratamento farmacológico , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Movimento Celular , Células Cultivadas , Proteína Rica em Cisteína 61 , DNA Complementar/metabolismo , Progressão da Doença , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Feminino , Fulvestranto , Biblioteca Gênica , Humanos , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Análise Multivariada , Transplante de Neoplasias , Regiões Promotoras Genéticas , Ligação Proteica , Receptores de Estrogênio/biossíntese , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tamoxifeno/farmacologia , Fatores de Tempo , Transfecção , Células Tumorais Cultivadas
12.
J Clin Oncol ; 19(6): 1649-57, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11250993

RESUMO

PURPOSE: To integrate stage, grade, and Eastern Cooperative Oncology Group (ECOG) performance status (PS) into a clinically useful tool capable of stratifying the survival of renal cell carcinoma (RCC) patients. PATIENTS AND METHODS: The medical records of 661 patients undergoing nephrectomy at University of California Los Angeles between 1989 and 1999 were evaluated. Median age was 61 years, male-to-female ratio was 2.2:1, and median follow-up was 37 months. Survival time was the primary end point assessed. Sixty-four possible combinations of stage, grade, and ECOG PS were analyzed and collapsed into distinct groups. The internal validity of the categorized was challenged by a univariate analysis and a multivariate analysis testing for the accountability of each UCLA Integrated Staging System (UISS) category against independent variables shown to have impact on survival. RESULTS: Combining and stratifying 1997 tumor-node-metastasis stage, Fuhrman's grade and ECOG PS resulted in five survival stratification groups designated UISS, and numbered I to V. The projected 2- and 5-year survival for the UISS groups are as follows for the groups: I, 96% and 94%; II, 89% and 67%; III, 66% and 39%; IV, 42% and 23%; and V, 9% and 0%, respectively. UISS accounted for the significant variables in the variate analysis. CONCLUSION: A novel system for staging and predicting survival for RCC integrating clinical variables is offered. UISS is simple to use and is superior to stage alone in differentiating patients' survival. Our data suggests that UISS is an important prognostic tool for counseling patients with various stages of kidney cancer. Further prospective large-scale validation with external data is awaited.


Assuntos
Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Estadiamento de Neoplasias/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Renais/classificação , Determinação de Ponto Final , Feminino , Nível de Saúde , Humanos , Neoplasias Renais/classificação , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Análise de Sobrevida
13.
Cancer Res ; 60(23): 6563-7, 2000 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-11118033

RESUMO

Prostatic small cell carcinoma is an aggressive subtype of prostate cancer that usually appears as a progression of the original adenocarcinoma. We describe here the WISH-PC2, a novel neuroendocrine xenograft of small cell carcinoma of the prostate. This xenograft was established from a poorly differentiated prostate adenocarcinoma and is serially transplanted in immune-compromised mice where it grows within the prostate, liver, and bone, inducing osteolytic lesions with foci of osteoblastic activity. It secretes to the mouse Chromogranin A and expresses prostate plasma carcinoma tumor antigen-1, six-transmembrane epithelial antigen of the prostate, and members of the Erb-B receptor family. It does not express prostate-specific antigen, prostate stem cell antigen, prostate-specific membrane antigen, and androgen receptor, and it grows independently of androgen. Altogether, WISH-PC2 provides an unlimited source in which to study the involvement of neuroendocrine cells in the progression of prostatic adenocarcinoma and can serve as a novel model for the testing of new therapeutic strategies for prostatic small cell carcinoma.


Assuntos
Carcinoma de Células Pequenas/patologia , Modelos Animais de Doenças , Neoplasias da Próstata/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Adenocarcinoma/patologia , Idoso , Animais , Carcinoma de Células Pequenas/tratamento farmacológico , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Fenótipo , Neoplasias da Próstata/tratamento farmacológico , Células Tumorais Cultivadas
14.
Clin Cancer Res ; 6(11): 4226-33, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11106236

RESUMO

Human herpesvirus 8 (HHV-8), also known as Kaposi's sarcoma-associated herpesvirus, has been implicated in the pathogenesis of Kaposi's sarcoma (KS), primary effusion lymphoma (PEL), multicentric Castleman's disease, and recently multiple myeloma (MM). DNA sequence analyses of HHV-8 suggest that multiple HHV-8 strains exist. We extracted DNA from 24 patients with MM and 3 patients with monoclonal gammopathy of undetermined significance and compared HHV-8 open reading frames (ORFs) 26 and 65 sequences with those derived from patients with KS, PEL, and two HHV-8-positive PEL cell lines KS-1 and BC-1. ORF26 sequence data suggest that MM patients are consistently carriers of HHV-8 strain subtype C3. All MM patients also consistently revealed either a single bp deletion or substitution at position 112197 in ORF65. This unique alteration is not present in patients with KS or PEL or in PEL cell lines. It occurs in the portion of ORF65 that is known to be responsible for a serological response to HHV-8.


Assuntos
Herpesvirus Humano 8/genética , Linfoma/virologia , Mieloma Múltiplo/virologia , Fases de Leitura Aberta , Sarcoma de Kaposi/virologia , Sequência de Aminoácidos , Sequência de Bases , Humanos , Dados de Sequência Molecular
15.
Bone Marrow Transplant ; 26(9): 1021-4, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11100284

RESUMO

Post-transplant lymphoproliferative disorder (PTLD) is a complication of allogeneic bone marrow transplantation (BMT). Rare cases of PTLD after autologous BMT have been reported only in adults. This case report is the first to describe PTLD in a pediatric patient after autologous peripheral stem cell transplantation (PSCT). This 2-year-old male with stage IV neuroblastoma underwent autologous PSCT. The post-PSCT course was complicated by fever with hematochezia and a lung mass. On day 94 post PSCT, colonoscopy revealed an ulcer due to a PTLD, monomorphic type, B cell phenotype, associated with Epstein-Barr virus. Fine needle aspiration identified the lung mass as neuroblastoma. PTLD can occur in pediatric autologous PSCT recipients, and may occur more frequently in autologous grafts manipulated by T cell depletion or CD34+ cell selection.


Assuntos
Neoplasias das Glândulas Suprarrenais/terapia , Transplante de Células-Tronco Hematopoéticas , Imunossupressores/efeitos adversos , Linfoma Difuso de Grandes Células B/etiologia , Neuroblastoma/terapia , Condicionamento Pré-Transplante/efeitos adversos , Neoplasias das Glândulas Suprarrenais/tratamento farmacológico , Neoplasias das Glândulas Suprarrenais/cirurgia , Adrenalectomia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Pré-Escolar , Cisplatino/administração & dosagem , Doenças do Colo/etiologia , Doenças do Colo/virologia , Terapia Combinada , Ciclofosfamida/administração & dosagem , Infecções por Citomegalovirus/etiologia , Doxorrubicina/administração & dosagem , Úlcera Duodenal/etiologia , Úlcera Duodenal/virologia , Infecções por Vírus Epstein-Barr/complicações , Etoposídeo/administração & dosagem , Hemorragia Gastrointestinal/etiologia , Humanos , Hospedeiro Imunocomprometido , Neoplasias Pulmonares/secundário , Metástase Linfática , Linfoma Difuso de Grandes Células B/patologia , Linfoma Difuso de Grandes Células B/virologia , Masculino , Neuroblastoma/tratamento farmacológico , Neuroblastoma/secundário , Neuroblastoma/cirurgia , Neoplasias Orbitárias/secundário , Doenças Priônicas , Transplante Autólogo , Úlcera/etiologia , Úlcera/virologia , Vincristina/administração & dosagem
16.
J Biol Chem ; 275(22): 16618-25, 2000 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-10748106

RESUMO

In most cells, transferrin receptor (TfR1)-mediated endocytosis is a major pathway for cellular iron uptake. We recently cloned the human transferrin receptor 2 (TfR2) gene, which encodes a second receptor for transferrin (Kawabata, H., Yang, R., Hirama, T., Vuong, P. T., Kawano, S., Gombart, A. F., and Koeffler, H. P. (1999) J. Biol. Chem. 274, 20826-20832). In the present study, the regulation of TfR2 expression and function was investigated. A select Chinese hamster ovary (CHO)-TRVb cell line that does not express either TfR1 or TfR2 was stably transfected with either TfR1 or TfR2-alpha cDNA. TfR2-alpha-expressing cells had considerably lower affinity for holotransferrin when compared with TfR1-expressing CHO cells. Interestingly, in contrast to TfR1, expression of TfR2 mRNA in K562 cells was not up-regulated by desferrioxamine (DFO), a cell membrane-permeable iron chelator. In MG63 cells, expression of TfR2 mRNA was regulated in the cell cycle with the highest expression in late G(1) phase and no expression in G(0)/G(1). DFO reduced cell proliferation and DNA synthesis of CHO-TRVb control cells, whereas it had little effect on TfR2-alpha-expressing CHO cells when measured by clonogenic and cell cycle analysis. In addition, CHO cells that express TfR2-alpha developed into tumors in nude mice whereas CHO control cells did not. In conclusion, TfR2 expression may be regulated by the cell cycle rather than cellular iron status and may support cell growth both in vitro and in vivo.


Assuntos
Divisão Celular/fisiologia , Quelantes de Ferro/química , Receptores da Transferrina/fisiologia , Animais , Células CHO , Ciclo Celular , Cricetinae , Humanos , Camundongos , RNA Mensageiro/genética , Receptores da Transferrina/genética , Transfecção
17.
Oncogene ; 19(10): 1288-96, 2000 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-10713670

RESUMO

Prostate stem cell antigen (PSCA) is a recently defined homologue of the Thy-1/Ly-6 family of glycosylphosphatidylinositol (GPI)-anchored cell surface antigens. PSCA mRNA is expressed in the basal cells of normal prostate and in more than 80% of prostate cancers. The purpose of the present study was to examine PSCA protein expression in clinical specimens of human prostate cancer. Five monoclonal antibodies were raised against a PSCA-GST fusion protein and screened for their ability to recognize PSCA on the cell surface of human prostate cancer cells. Immunohistochemical analysis of PSCA expression was performed on paraffin-embedded sections from 25 normal tissues, 112 primary prostate cancers and nine prostate cancers metastatic to bone. The level of PSCA expression in prostate tumors was quantified and compared with expression in adjacent normal glands. The antibodies detect PSCA expression on the cell surface of normal and malignant prostate cells and distinguish three extracellular epitopes on PSCA. Prostate and transitional epithelium reacted strongly with PSCA. PSCA staining was also seen in placental trophoblasts, renal collecting ducts and neuroendocrine cells in the stomach and colon. All other normal tissues tested were negative. PSCA protein expression was identified in 105/112 (94%) primary prostate tumors and 9/9 (100%) bone metastases. The level of PSCA expression increased with higher Gleason score (P=0.016), higher tumor stage (P=0.010) and progression to androgen-independence (P=0. 021). Intense, homogeneous staining was seen in all nine bone metastases. PSCA is a cell surface protein with limited expression in extraprostatic normal tissues. PSCA expression correlates with tumor stage, grade and androgen independence and may have prognostic utility. Because expression on the surface of prostate cancer cells increases with tumor progression, PSCA may be a useful molecular target in advanced prostate cancer.


Assuntos
Antígenos de Neoplasias/isolamento & purificação , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/secundário , Glicoproteínas de Membrana/isolamento & purificação , Proteínas de Neoplasias/isolamento & purificação , Neoplasias da Próstata/imunologia , Anticorpos Monoclonais , Anticorpos Antineoplásicos , Antígenos de Neoplasias/imunologia , Sistema Digestório/imunologia , Epitopos , Proteínas Ligadas por GPI , Humanos , Túbulos Renais Coletores/imunologia , Masculino , Glicoproteínas de Membrana/imunologia , Proteínas de Neoplasias/imunologia , Estadiamento de Neoplasias , Sistemas Neurossecretores/imunologia , Neoplasias da Próstata/patologia , Distribuição Tecidual , Trofoblastos/imunologia
18.
Leuk Res ; 24(1): 59-61, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10634647

RESUMO

The presence and distribution of Epstein-Barr virus (EBV), as well as human herpesvirus-6 and-8 (HHV-6 and HHV-8) was investigated by polymerase chain reaction in 191 samples from a variety of lymphoproliferative disorders. HHV-6 DNA was detected in 18% (30 of 169) of non-HHV-8 related lymphoproliferative disorders, with the highest frequency in AIDS-related lymphomas (8 of 25, 32%). In contrast, HHV-6 DNA was present in less than 5% (1 of 22) of HHV-8 related lymphoproliferative disorders [21 primary effusion lymphomas (PEL), and 1 cases of Castleman disease]. As compared to HHV-6, EBV DNA was frequently detected in PEL (11 of 19 samples, 58%). This study suggests that transformation to PEL is not enhanced by HHV-6, furthermore HHV-6 and -8 may interfere with each other.


Assuntos
Infecções por Herpesviridae/epidemiologia , Herpesvirus Humano 6/isolamento & purificação , Herpesvirus Humano 8/isolamento & purificação , Linfoma de Células B/virologia , Infecções Tumorais por Vírus/epidemiologia , Hiperplasia do Linfonodo Gigante/epidemiologia , Hiperplasia do Linfonodo Gigante/virologia , Transformação Celular Neoplásica , Transformação Celular Viral , Comorbidade , DNA Viral/isolamento & purificação , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/epidemiologia , Infecções por Herpesviridae/complicações , Herpesvirus Humano 4/isolamento & purificação , Herpesvirus Humano 4/patogenicidade , Herpesvirus Humano 6/patogenicidade , Herpesvirus Humano 8/patogenicidade , Humanos , Japão/epidemiologia , Linfoma Relacionado a AIDS/epidemiologia , Linfoma Relacionado a AIDS/virologia , Linfoma de Células B/epidemiologia , Linfoma de Zona Marginal Tipo Células B/epidemiologia , Linfoma de Zona Marginal Tipo Células B/virologia , Linfoma não Hodgkin/epidemiologia , Linfoma não Hodgkin/virologia , Transtornos Linfoproliferativos/epidemiologia , Transtornos Linfoproliferativos/virologia , Reação em Cadeia da Polimerase , Prevalência , Infecções Tumorais por Vírus/complicações , Interferência Viral
19.
Br J Cancer ; 82(2): 452-8, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10646904

RESUMO

The organic arsenical known as melarsoprol (Mel-B) is used to treat African trypanosomiasis. Recently, another arsenical, As2O3 was shown to be effective in treatment of acute promyelocytic leukaemia. We have investigated the anti-tumour activities of Mel-B either with or without all-trans-retinoic acid (ATRA) using the MCF-7 human breast cancer cells, as well as the PC-3 and DU 145 human prostate cancer cells both in vitro and in vivo. The antiproliferative effects of Mel-B and/or ATRA against breast and prostate cancer were tested in vitro using clonogenic assays and in vivo in triple immunodeficient mice. Furthermore, the mechanism of action of these compounds was studied by examining the cell cycle, levels of bcl-2, apoptosis and antiproliferative potency using a pulse-exposure assay. Clonogenic assays showed that the cancer cell lines were sensitive to the inhibitory effect of Mel-B (effective dose that inhibited 50% clonal growth [ED50]: 7 x 10(-9) M for MCF-7, 2 x 10(-7) M for PC-3, 3 x 10(-7) M for DU145 cells. Remarkably, the combination of Mel-B and ATRA had an enhanced antiproliferative activity against all three cancer cell lines. Furthermore, the combination of Mel-B and ATRA induced a high level of apoptosis in all three cell lines. Treatment of PC-3 and MCF-7 tumours growing in triple immunodeficient mice with Mel-B and ATRA either alone or in combination markedly retarded tumour size and weight of the tumours without major side-effects. In conclusion, our results suggest that either Mel-B alone or with ATRA may be a useful, novel therapy for breast and prostate cancers.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Melarsoprol/farmacologia , Neoplasias da Próstata/patologia , Tretinoína/farmacologia , Tripanossomicidas/farmacologia , Animais , Apoptose , Neoplasias da Mama/tratamento farmacológico , Divisão Celular/efeitos dos fármacos , Feminino , Humanos , Masculino , Camundongos , Neoplasias da Próstata/tratamento farmacológico , Células Tumorais Cultivadas
20.
Prostate ; 42(3): 163-71, 2000 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10639186

RESUMO

BACKGROUND: Management of prostate cancer that has spread beyond the capsule is a difficult problem. Innovative and nontoxic approaches to the disease are urgently required. Recently, a commercially available herbal mixture called PC-SPES showed potent antitumor activities on a variety of malignant cells in vitro. METHODS: PC-SPES was evaluated for its ability to inhibit clonal growth, and to induce cell cycle arrest of three human prostate cancer cell lines (LNCaP, PC-3, and DU 145). Western blot analysis examined the effect of PC-SPES on levels of p21(waf1), p27(kip1), Bcl-2, and E-cadherin in the three cell lines; and telomerase activity was examined by telomeric repeat amplification protocol (TRAP) assay. Furthermore, the effect of oral PC-SPES (250 mg/kg/day) on growth of PC-3 and DU 145 tumors present in male BNX nu/nu triple immunodeficient mice was studied. LNCaP cells were not analyzed in mice because they grow only with difficulty in these immunodeficient mice. RESULTS: PC-SPES markedly inhibited clonal growth of LNCaP, PC-3, and DU 145 prostate cancer cells, with a 50% inhibition (ED50) at approximately 2 microl/ml. Pulse-exposure studies showed that a 5-day pulse-exposure to PC-SPES (2 microl/ml) in liquid culture achieved a 50% inhibition of PC-3 clonal growth in soft agar, suggesting that the growth inhibition mediated by the extracts remained after removal of PC-SPES. Cell cycle analysis using the prostate cancer cell lines found that PC-SPES induced a significant increase in the number of cells in G0-G1 and G2/M, with a concomitant decrease in the number of cells in S phase. PC-SPES (2 microl/ml, 4 days) increased slightly the levels of p21(waf1) in the three cell lines, decreased by 40% the levels of Bcl-2 in PC-3, and the levels of p27(kip1) and E-cadherin and telomerase were unchanged in each of the lines. In vivo treatment with oral PC-SPES of male BNX mice having DU 145 tumors produced significant inhibition of their growth (P < 0.001), with no objective side effects including blood chemistries, weights, or autopsy analysis. The PC-SPES showed no statistical effect on the in vivo growth of PC-3 cells. CONCLUSIONS: PC-SPES inhibits clonal proliferation of human prostate cancer cells both in vitro and in vivo, using a murine model.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Medicamentos de Ervas Chinesas , Extratos Vegetais/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Androgênios/metabolismo , Animais , Antineoplásicos Fitogênicos/farmacologia , Caderinas/metabolismo , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Quinases Ciclina-Dependentes/antagonistas & inibidores , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Extratos Vegetais/farmacologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Telomerase/metabolismo , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...