Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Clin Oncol ; 21(11): 2085-93, 2003 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12775733

RESUMO

PURPOSE: Cyclin D1 is overexpressed in almost 60% of resectable non-small-cell lung cancer (NSCLC). In the absence of cyclin D1 gene amplification, overexpression is characterized by allelic imbalanced transcript levels. METHODS: The aims were to study cyclin D1 expression by immunohistochemistry and allelic balance of transcripts in tumor-free bronchial epithelia from patients with resectable NSCLC by using monoclonal antibodies (48 patients and 288 sites), microdissection/reverse transcriptase polymerase chain reaction/restriction fragment length polymorphism analyses (24 patients and 144 sites). Derived data were related to patient characteristics-in particular, smoking habits. RESULTS: In 167 (58%) of 288 sites, cyclin D1 was overexpressed, with cytoplasmic and nuclear sublocalization in 53% and 7% of all sites, respectively. Nuclear overexpression was more frequent in premalignant versus normal or hyperplastic epithelia (55% v 3%; P <.0001). Allele-specific expression imbalances were found in 69 (48%) of 144 sites; in particular, those in which cyclin D1 was overexpressed (P =.004). In 14 (58%) of 24 patients, balanced or imbalanced transcript ratios and degree of expression were consistent at all sites for the same patient, whereas in another 10 patients, transcript balances and cyclin D1 expression patterns varied across the sites. Nuclear cyclin D1 expression in at least one site (14 of 48 patients) was linked to heavy smoking (> 40 pack-years; P =.02) and shorter overall survival (P =.01). CONCLUSION: Allele-specific, probably damage-driven, deregulation of the cyclin D1 gene may precede and perhaps facilitate the spread of preneoplastic clones across the bronchial epithelial surface in a significant number of patients. Cyclin D1 expression at multiple bronchial sites may identify a subgroup of heavy-smoking patients with poor outcome.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Ciclina D1/biossíntese , Ciclina D1/genética , Neoplasias Pulmonares/metabolismo , Mucosa Respiratória/metabolismo , Fumar/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Polimorfismo de Fragmento de Restrição , Modelos de Riscos Proporcionais , Estudos Prospectivos , Estatísticas não Paramétricas , Taxa de Sobrevida , Suíça/epidemiologia
2.
Cell Death Differ ; 9(9): 905-14, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12181741

RESUMO

Caspase-3 activity has been described to be essential for drug-induced apoptosis. Recent results suggest that in addition to its downstream executor function, caspase-3 is also involved in the processing of upstream caspase-8 and -9. To test the absolute requirement for caspase-3, we examined mitomycin C (MMC)-induced apoptosis in the caspase-3 deficient human breast cancer cell line MCF-7. MMC was used as anticancer drug since this agent was preferentially active compared to chemotherapeutic compounds with differing mechanisms of action such as cisplatin, docetaxel, or lovastatin. MMC treatment led to pronounced caspase-8, -9, and -7 processing and early morphological features of apoptosis within 48 h. This could be inhibited by the broad-spectrum caspase inhibitor z-VAD.fmk and to a lesser extent by z-IETD.fmk and z-LEHD.fmk, which have a certain preference for inhibiting caspase-8 and -9, respectively. MMC induced apoptosis in MCF-7 cells was not mediated by the death receptor pathway as demonstrated by experiments using the inhibiting anti-Fas antibody ZB4 and transfections with CrmA, a viral serpin inhibitor of caspase-8, and the dominant negative Fas-associated death domain (FADD-DN). Stable expression with Bcl-2 significantly prevented the processing of caspase-9 but also of caspase-8 and blocked the induction of apoptosis. Thus, we provide evidence that caspase-3 activity is dispensable for MMC-induced apoptosis and for caspase-8 and -9 processing in MCF-7 cells.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Apoptose/fisiologia , Neoplasias da Mama/enzimologia , Carcinoma/enzimologia , Caspases/metabolismo , Mitomicina/farmacologia , Receptor fas/metabolismo , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Carcinoma/tratamento farmacológico , Caspase 3 , Caspase 7 , Caspase 8 , Caspase 9 , Caspases/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas , Receptor fas/efeitos dos fármacos
3.
Cell Death Differ ; 8(12): 1213-23, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11753569

RESUMO

Inactivation of the tumour suppressor p53 is the most common defect in cancer cells. p53 is a sequence specific transcription factor that is activated in response to various forms of genotoxic stress to induce cell cycle arrest and apoptosis. Induction of p53 is subjected to complex and strict control through several pathways, as it will often determine cellular fate. The p73 protein shares strong structural and functional similarities with p53 such as the potential to activate p53 responsive genes and the ability to induce apoptosis. In addition to alternative splicing at the carboxyl terminus which yields several p73 isoforms, a p73 variant lacking the N-terminal transactivation domain (Delta Np73) was described in mice. In this study, we report the cloning and characterisation of the human Delta Np73 isoforms, their regulation by p53 and their possible role in carcinogenesis. As in mice, human Delta Np73 lacks the transactivation domain and starts with an alternative exon (exon 3'). Its expression is driven by a second promoter located in a genomic region upstream of this exon, supporting the idea of two independently regulated proteins, derived from the same gene. As anticipated, Delta Np73 is capable of regulating TAp73 and p53 function since it is able to block their transactivation activity and their ability to induce apoptosis. Interestingly, expression of the Delta Np73 is strongly up-regulated by the TA isoforms and by p53, thus creating a feedback loop that tightly regulates the function of TAp73 and more importantly of p53. The regulation of Delta Np73 is exerted through a p53 responsive element located on the Delta N promoter. Expression of Delta Np73 not only regulates the function of p53 and TAp73 but also shuts off its own expression, once again finely regulating the whole system. Our data also suggest that increased expression of Delta Np73, functionally inactivating p53, could be involved in tumorogenesis. An extensive analysis of the expression pattern of Delta Np73 in primary tumours would clarify this issue.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Genes p53 , Proteínas Nucleares/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem de Organismos , Proteínas de Ligação a DNA/genética , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Genes Supressores de Tumor , Humanos , Íntrons , Camundongos , Dados de Sequência Molecular , Proteínas Nucleares/genética , Fosfoproteínas Fosfatases , Regiões Promotoras Genéticas , Ativação Transcricional/genética , Ativação Transcricional/fisiologia , Transfecção , Células Tumorais Cultivadas , Proteína Tumoral p73 , Proteínas Supressoras de Tumor
5.
Oncogene ; 19(52): 6082-90, 2000 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-11146561

RESUMO

To develop a new approach to the treatment of primitive neuroectodermal tumors we evaluated the effect of the HMG-CoA reductase inhibitor lovastatin on the Ewing's sarcoma cell line CHP-100. Lovastatin induced neural morphology and markers including neuron-specific enolase and neurofilament protein. The acquisition of neural morphology required new mRNA synthesis, and cDNA microarray analysis confirmed that lovastatin altered the program of gene expression. After morphologic differentiation the cells underwent rapidly progressive apoptosis. In normal development of neuronal progenitors, differentiation signals trigger p21WAF1 accumulation, RB hypophosphorylation, enhanced RB-E2F-1 association, and G1 arrest, and these events have been shown to protect from apoptosis. In contrast, in the Ewing's sarcoma cells lovastatin triggered differentiation without causing cell cycle arrest: p21WAF1 was not induced, RB remained hyperphosphorylated, and RB protein expression and RB-E2F-1 association were markedly downregulated, suggesting that loss of an RB-regulated G1 checkpoint promoted apoptosis. Consistent with this hypothesis, adenoviral p21WAF1 decreased DNA synthesis and partially protected from lovastatin-induced cytotoxicity. The data demonstrate a new model for examining the genetic regulation of cell fate in a neural progenitor tumor and suggest a new approach to the treatment of this neoplasm.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular , Ectoderma/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Lovastatina/farmacologia , Neurônios/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo , Proteínas Supressoras de Tumor , Biomarcadores/análise , Tamanho Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21 , Inibidor de Quinase Dependente de Ciclina p27 , Ciclinas/metabolismo , DNA/biossíntese , Regulação para Baixo/efeitos dos fármacos , Ectoderma/patologia , Ectoderma/ultraestrutura , Citometria de Fluxo , Genes cdc , Humanos , Imuno-Histoquímica , Microscopia Eletrônica de Varredura , Proteínas Associadas aos Microtúbulos/metabolismo , Índice Mitótico , Neurônios/patologia , Neurônios/ultraestrutura , Sarcoma de Ewing/patologia , Sarcoma de Ewing/ultraestrutura , Células Tumorais Cultivadas
6.
J Biol Chem ; 273(17): 10618-23, 1998 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-9553123

RESUMO

Progression through the cell cycle is controlled by the induction of cyclins and the activation of cognate cyclin-dependent kinases. The 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitor lovastatin induces growth arrest and cell death in certain cancer cell types. We have pursued the mechanism of growth arrest in PC-3-M cells, a p53-null human prostate carcinoma cell line. Lovastatin treatment increased protein and mRNA levels of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1), increased binding of p21 with Cdk2, markedly inhibited cyclin E- and Cdk2-associated phosphorylation of histone H1 or GST-retinoblastoma protein, enhanced binding of the retinoblastoma protein to the transcription factor E2F-1 in vivo, and induced the activation of a p21 promoter reporter construct. By using p21 promoter deletion constructs, the lovastatin-responsive element was mapped to a region between -93 and -64 relative to the transcription start site. Promoter mutation analysis indicated that the lovastatin-responsive site coincided with the previously identified transforming growth factor-beta-responsive element. These data indicate that in human prostate carcinoma cells an inhibitor of the HMG-CoA reductase pathway can circumvent the loss of wild-type p53 function and induce critical downstream regulatory events leading to transcriptional activation of p21.


Assuntos
Proteínas de Transporte , Proteínas de Ciclo Celular , Ciclinas/genética , Proteínas de Ligação a DNA , Regulação Neoplásica da Expressão Gênica , Hidroximetilglutaril-CoA Redutases/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Neoplasias da Próstata/genética , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Apoptose/efeitos dos fármacos , Sequência de Bases , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Fatores de Transcrição E2F , Fator de Transcrição E2F1 , Fase G1/efeitos dos fármacos , Humanos , Lovastatina/farmacologia , Masculino , Dados de Sequência Molecular , Fosforilação , Regiões Promotoras Genéticas , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteína do Retinoblastoma/metabolismo , Proteína 1 de Ligação ao Retinoblastoma , Fator de Transcrição DP1 , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas
7.
Biochem Pharmacol ; 48(11): 2117-26, 1994 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-7528507

RESUMO

We examined the cytotoxicity, biochemical effects and metabolism of 4-methoxy-8-(beta-D-ribofuranosylamino)pyrimido[5,4-d]pyrimidine (MRPP), a synthetic nucleoside inhibitor of phosphoribosylpyrophosphate synthetase, in HCT 116 human colorectal cancer cells. A 4-hr exposure to 1 and 10 microM MRPP inhibited cell growth over a 72-hr period by 76 and 89%, and inhibited clonogenic capacity by 36 and 65%, respectively. MRPP was avidly metabolized to the 5'-monophosphate derivative (MRPP-MP), and MRPP-MP formation increased with increasing MRPP exposure (microM.hr). MRPP-MP was stable, and the intracellular half-life was in excess of 48 hr. A 4-hr exposure to 10 microM MRPP resulted in significant decreases in ATP, UTP, GTP, CTP, dATP, dTTP, and PRPP pools. Near maximal ribonucleotide triphosphate depletion was achieved with > or = 24 microM.hr MRPP, and growth inhibition as a function of MRPP microM.hr closely reflected the biochemical effects. Ribonucleotide triphosphate pools remained depleted for up to 48 hr after drug removal, apparently as a consequence of the prolonged retention of MRPP-MP. MRPP (10 microM) inhibited the salvage of [3H]guanine, [3H]adenine and [3H]guanosine, and concurrent exposure to MRPP and either 100 microM adenine, hypoxanthine, or guanine did not reverse ATP or GTP depletion. Concurrent exposure to 10 microM MRPP and either 10 microM adenosine, uridine or thymidine was accompanied by repletion of ATP, UTP, and dTTP pools, respectively, but depletion of other nucleotide pools was not corrected. In contrast, 10 microM guanosine did not correct GTP depletion in the presence of MRPP. The combination of 10 microM each of thymidine, uridine, adenosine and guanosine during and following a 24-hr exposure to MRPP provided partial protection against 0.1 or 1 microM MRPP, but did not affect the cytotoxicity associated with 10 microM MRPP. MRPP is a novel antimetabolite that inhibits both de novo and salvage pathways for purine synthesis and de novo pyrimidine synthesis.


Assuntos
Antimetabólitos/toxicidade , Nucleosídeos de Pirimidina/toxicidade , Trifosfato de Adenosina/metabolismo , Antimetabólitos/metabolismo , Linhagem Celular , Citidina Trifosfato/metabolismo , DNA/biossíntese , DNA/isolamento & purificação , Humanos , Concentração de Íons de Hidrogênio , Nucleosídeos/farmacologia , Fosforilação , Nucleosídeos de Pirimidina/metabolismo , RNA/biossíntese , Uridina Trifosfato/metabolismo
8.
FEBS Lett ; 353(2): 129-32, 1994 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-7926036

RESUMO

The detergent Triton X-100 (TX100) was used with the intention to establish a model for necrotic cell death. However, TX100 was found to induce apoptotic and necrotic death in prostate and colon cancer cell lines. Apoptosis was characterized by the typical morphological features and internucleosomal DNA fragmentation. The rapid onset within 60 min and the lack of inhibition by cycloheximide indicated that apoptosis induced by TX100 was not dependent on protein synthesis. Removal of extracellular calcium blocked internucleosomal DNA fragmentation. This pattern of cell death shows a striking similarity to the effect of cytotoxic lymphocytes on their target cells.


Assuntos
Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Octoxinol/farmacologia , Linfócitos T Citotóxicos/fisiologia , Cálcio/metabolismo , Neoplasias do Colo/patologia , Cicloeximida/farmacologia , DNA de Neoplasias/efeitos dos fármacos , Humanos , Cinética , Masculino , Neoplasias da Próstata/patologia , Biossíntese de Proteínas , Células Tumorais Cultivadas
9.
Proc Natl Acad Sci U S A ; 91(12): 5330-4, 1994 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-8202489

RESUMO

Recent clinicopathologic studies have shown that many prostatic adenocarcinomas express focal neuroendocrine differentiation and that neuroendocrine differentiation is most apparent in advanced anaplastic tumors. While studying growth-regulatory signal transduction events in human prostate carcinoma cell lines, we found that in two of four cell lines, the androgen-sensitive line LNCaP and the highly metastatic androgen-independent line PC-3-M, elevation of cAMP through addition of cAMP analogues or phosphodiesterase inhibitors induced a markedly neuronal morphology. Also in LNCaP cells ultrastructural analysis showed that cAMP induced the appearance of neurosecretory cell-like dense-core granules. Phenotypic analysis of untreated LNCaP and PC-3-M cells showed that both cell lines express markers of the neural crest including S-100, chromogranin A, pp60c-src, and neuron-specific enolase as well as the epithelial marker KS1/4 and stage-specific embryonic antigen 4. In PC-3-M cells, cAMP markedly elevated neuron-specific enolase protein and caused an increase in the specific activity of the neuroendocrine marker pp60c-src, and in both cell lines expression of KS1/4 and stage-specific embryonic antigen 4 was down-regulated. In addition to effects on lineage markers, cAMP treatment induced G1 synchronization, growth arrest, and loss of clonogenicity, indicating terminal differentiation. Our data provide direct evidence of plasticity in the lineage commitment of adenocarcinoma of the prostate. We have shown that cell-permeant cAMP analogues can induce terminal differentiation, suggesting that hydrolysis-resistant cyclic nucleotides may present an additional approach to the treatment of advanced prostate cancer.


Assuntos
Adenocarcinoma/patologia , AMP Cíclico/fisiologia , Neoplasias da Próstata/patologia , Diferenciação Celular , Divisão Celular , Linhagem Celular , Cromogranina A , Cromograninas/metabolismo , Citoplasma/fisiologia , Glândulas Endócrinas/citologia , Células Epiteliais , Humanos , Masculino , Neurônios/citologia , Fosfopiruvato Hidratase/metabolismo , Proteínas S100/metabolismo
10.
J Clin Invest ; 89(1): 191-6, 1992 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1309535

RESUMO

To develop a new approach to the treatment of advanced, hormone-refractory prostate cancer, the signal transductions regulating the growth of human androgen-independent prostate carcinoma cell lines were studied. Agonist-stimulated Ca2+ mobilization, a critical regulatory event in other secretory cell types, was studied as a means of identifying previously undescribed plasma membrane receptors that may transduce a growth inhibitory signal. In all of the cell lines tested, P2-purinergic receptor agonists, including ATP and certain hydrolysis-resistant adenine nucleotides, induced a rapid, transient increase in cytoplasmic free Ca2+ that was detectable at 50 to 100 nM ATP, was maximal at 100 microM ATP, and was inhibited approximately 50% by chelation of extracellular Ca2+. Within 8 s after addition, ATP stimulated accumulation of the polyphosphatidylinositol products inositol (1, 4, 5) trisphosphate, inositol (1, 3, 4) trisphosphate, and inositol tetrakisphosphate. In addition to stimulating phosphatidylinositol turnover and Ca2+ mobilization, ATP and hydrolysis-resistant ATP analogues induced greater than 90% inhibition of the growth of all lines tested. These data demonstrate that human androgen-independent prostate carcinoma cells express functional P2-purinergic receptors linked to phospholipase C, and that agonists of this receptor are markedly growth inhibitory, suggesting a novel therapeutic approach to this common adult neoplasm.


Assuntos
Androgênios/metabolismo , Carcinoma/metabolismo , Neoplasias da Próstata/metabolismo , Receptores Purinérgicos/metabolismo , Nucleotídeos de Adenina/farmacologia , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Estudos de Avaliação como Assunto , Humanos , Fosfatos de Inositol/metabolismo , Masculino , Fosfatidilinositóis/metabolismo , Transdução de Sinais/fisiologia , Células Tumorais Cultivadas , Fosfolipases Tipo C/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA